EP3130605A1 - Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers - Google Patents

Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers Download PDF

Info

Publication number
EP3130605A1
EP3130605A1 EP16186634.8A EP16186634A EP3130605A1 EP 3130605 A1 EP3130605 A1 EP 3130605A1 EP 16186634 A EP16186634 A EP 16186634A EP 3130605 A1 EP3130605 A1 EP 3130605A1
Authority
EP
European Patent Office
Prior art keywords
frα
antibody
seq
cancer
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP16186634.8A
Other languages
German (de)
French (fr)
Other versions
EP3130605B1 (en
Inventor
Daniel J. O'shannessy
Luigi Grasso
Shanhong Wan
Qimin Chao
Elizabeth Brooke Somers
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eisai Inc
Original Assignee
Morphotek Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Morphotek Inc filed Critical Morphotek Inc
Publication of EP3130605A1 publication Critical patent/EP3130605A1/en
Application granted granted Critical
Publication of EP3130605B1 publication Critical patent/EP3130605B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57488Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds identifable in body fluids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57449Specifically defined cancers of ovaries
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/82Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving vitamins or their receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/56Staging of a disease; Further complications associated with the disease

Definitions

  • alpha, beta, and gamma the high affinity receptor for folate comes in three isoforms: alpha, beta, and gamma.
  • the alpha and beta forms are typically bound to the membranes of cells by a glycosyl phosphatidylinositol (GPI) anchor. They recycle between extracellular and endocytic compartments and are capable of transporting folate into the cell.
  • Soluble forms of FR ⁇ may be derived by the action of proteases or phospholipase on membrane anchored folate receptors.
  • Folate receptor alpha (also referred to as FR ⁇ , FR-alpha, FOLR-1 or FOLR1) is expressed in a variety of epithelial tissues, including those of the choroid plexus, lung, thyroid, kidney, uterus, breast, Fallopian tube, epididymis, and salivary glands.
  • FR ⁇ FR ⁇
  • FR-alpha FR-alpha
  • FOLR-1 FOLR-1
  • FR ⁇ Folate receptor alpha
  • lung cancer e.g., bronchioalveolar carcinomas, carcinoid tumors, and non-small cell lung cancers, such as adenocarcinomas
  • mesothelioma ovarian cancer
  • renal cancer e.g., brain cancer (e.g., anaplastic ependymoma, cerebellar juvenile pilocytic astrocytoma, and brain metastases); cervical cancer; nasopharyngeal cancer; mesodermally derived tumor; squamous cell carcinoma of the head and neck; endometrial cancer; endometrioid adenocarcinomas of the ovary, serous cystadenocarcinomas, breast cancer; bladder cancer; pancreatic cancer; bone cancer ( e.g., high-grade osteosarcoma); pituitary cancer (e.g., pituitary adenomas); colorectal cancer and medullary thyroid cancer.
  • lung cancer e.g.,
  • FR ⁇ expression is associated with a worse prognosis
  • other types of cancers e.g., non-small-cell lung cancers
  • FR ⁇ expression is associated with a better prognosis. See, e.g., Iwakiri S et al. Annals of Surgical Oncology, 15(3): 889-899 ; Saba N.F. et al. Head Neck, 31(4): 475-481 (2009 ).
  • the present invention provides methods of assessing whether a subject is afflicted with FR ⁇ -expressing cancers such as lung or ovarian cancer, methods of assessing the progression of FR ⁇ -expressing cancers such as lung or ovarian cancer in a subject afflicted with the FR ⁇ -expressing cancers, methods of stratifying an FR ⁇ -expressing cancer subject into one of at least four cancer therapy groups, methods of assessing the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer and kits for assessing whether a subject is afflicted with FR ⁇ -expressing cancers such as lung or ovarian cancer or for assessing the progression of FR ⁇ -expressing cancers such as lung or ovarian cancer in a subject.
  • the present invention provides a method of assessing whether a subject is afflicted with an FR ⁇ -expressing cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with an FR ⁇ -expressing cancer; wherein the level of FR ⁇ in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FR ⁇ .
  • the sample is either urine, serum, plasma or ascites.
  • the present invention is directed to a method of assessing whether a subject is afflicted with an FR ⁇ -expressing cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the urine sample derived from the subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the urine sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with an FR ⁇ -expressing cancer.
  • FR ⁇ folate receptor alpha
  • the present invention provides a method of assessing whether a subject is afflicted with a cancer that expresses FR ⁇ , by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the serum sample derived from the subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the serum sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with an FR ⁇ -expressing cancer.
  • FR ⁇ folate receptor alpha
  • the FR ⁇ -expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer.
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is non-small cell lung cancer, such as an adenocarcinoma.
  • the present invention is directed to methods of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FR ⁇ which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer.
  • FR ⁇ folate receptor alpha
  • the level of FR ⁇ is determined by contacting the sample with an antibody that binds FR ⁇ .
  • the antibody is selected from the group consisting of:
  • the antibody binds the same epitope as the MORAb-003 antibody.
  • the antibody includes SEQ ID NO: 1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • the antibody is the MOV18 antibody.
  • the antibody binds the same epitope as the MOV18 antibody.
  • the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21).
  • the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the level of FR ⁇ in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV 18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • a pair of antibodies selected from the group consisting of (a) MOV 18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody.
  • the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • the level of FR ⁇ is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • control sample is a standardized control level of FR ⁇ in a healthy subject.
  • the sample is treated with guanidine prior to determining the level of FR ⁇ in the sample.
  • the sample is diluted prior to determining the level of FR ⁇ in the sample.
  • the sample is centrifuged, vortexed, or both, prior to determining the level of FR ⁇ in the sample.
  • the present invention is directed to a method of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample with the level of FR ⁇ in a control sample, wherein a difference between the levels of FR ⁇ in the sample derived from the subject and in the control sample is an indication that the subject is afflicted with ovarian cancer; wherein the level of FR ⁇ in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobil
  • the present invention is directed to a method of assessing the progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the cancer will progress rapidly; and wherein a decrease in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the cancer will progress slowly or will regress, thereby assessing the progression of the FR ⁇ -expressing cancer in the subject; wherein the level of FR ⁇ which is not bound to a cell in the sample derived from the subject is assessed by contacting
  • the present invention provides a method of assessing the progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the urine sample derived from the subject with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the urine sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the cancer will progress rapidly; and wherein a decrease in the level of FR ⁇ in the urine sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the cancer will progress slowly or will regress, thereby assessing the progression of the FR ⁇ -expressing cancer in the subject.
  • FR ⁇ folate receptor alpha
  • the present invention provides methods of assessing the progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the serum sample derived from the subject with the level of FR ⁇ in control sample, wherein an increase in the level of FR ⁇ in the serum sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the cancer will progress rapidly; and wherein a decrease in the level of FR ⁇ in the serum sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the cancer will progress slowly or will regress, thereby assessing the progression of the FR ⁇ -expressing cancer in the subject.
  • FR ⁇ folate receptor alpha
  • the FR ⁇ -expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer.
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is non-small cell lung cancer, such as an adenocarcinoma.
  • the present invention is directed to methods of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FR ⁇ which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer.
  • FR ⁇ folate receptor alpha
  • the level of FR ⁇ is determined by contacting the sample with an antibody that binds FR ⁇ .
  • the antibody is selected from the group consisting of:
  • the antibody binds the same epitope as the MORAb-003 antibody.
  • the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • the antibody is the MOV 18 antibody.
  • the antibody binds the same epitope as the MOV18 antibody.
  • the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21).
  • the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the level of FR ⁇ in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody.
  • the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • the level of FR ⁇ is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • control sample is a standardized control level of FR ⁇ in a healthy subject.
  • control sample is a sample previously obtained from the subject.
  • the sample is treated with guanidine prior to determining the level of FR ⁇ in the sample.
  • the sample is diluted prior to determining the level of FR ⁇ in the sample.
  • the sample is centrifuged, vortexed, or both, prior to determining the level of FR ⁇ in the sample.
  • the present invention provides methods of assessing the progression of ovarian cancer in a subject afflicted with ovarian cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the ovarian cancer will progress rapidly; and wherein a decrease in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the ovarian cancer will progress slowly or will regress, thereby assessing the progression of ovarian cancer in the subject; wherein the level of FR ⁇ in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized
  • the present invention provides a method of stratifying a subject afflicted with an FR ⁇ -expressing cancer into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell; wherein the level of FR ⁇ which is not bound to a cell in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FR ⁇ .
  • the sample is selected from the group consisting of urine, serum, plasma or ascites.
  • the present invention provides a method of stratifying a subject afflicted with an FR ⁇ -expressing cancer into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample.
  • FR ⁇ folate receptor alpha
  • the present invention is directed to methods of stratifying a subject afflicted with an FR ⁇ -expressing cancer into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a serum sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the serum sample.
  • FR ⁇ folate receptor alpha
  • the FR ⁇ -expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer.
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is non-small cell lung cancer, such as an adenocarcinoma.
  • the present invention is directed to methods of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FR ⁇ which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer.
  • FR ⁇ folate receptor alpha
  • the level of FR ⁇ is determined by contacting the sample with an antibody that binds FR ⁇ .
  • the antibody is selected from the group consisting of:
  • the antibody binds the same epitope as the MORAb-003 antibody.
  • the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • the antibody is the MOV18 antibody.
  • the antibody binds the same epitope as the MOV 18 antibody.
  • the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21).
  • the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the level of FR ⁇ in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody.
  • the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • the level of FR ⁇ is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • control sample is a standardized control level of FR ⁇ in a healthy subject.
  • the sample is treated with guanidine prior to determining the level of FR ⁇ in the sample.
  • the sample is diluted prior to determining the level of FR ⁇ in the sample.
  • the sample is centrifuged, vortexed, or both, prior to determining the level of FR ⁇ in the sample.
  • the subject is stratified in Stage I, Stage II, Stage III or Stage IV ovarian cancer.
  • the present invention provides a method of stratifying an ovarian cancer subject into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample; wherein the level of FR ⁇ in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • the sample may be urine, serum, plasma or ascites.
  • the present invention provides a method of monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a sample derived from the subject, wherein the subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is efficacious.
  • the present invention provides a method of monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject, wherein the subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the urine sample derived from the subject with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the urine sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FR ⁇ in the urine sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is efficacious.
  • FR ⁇ folate receptor alpha
  • the present invention is directed to a method of monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a serum sample derived from the subject, wherein the subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the serum sample derived from the subject with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the serum sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FR ⁇ in the serum sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is efficacious.
  • FR ⁇ folate receptor alpha
  • the level of FR ⁇ is determined by contacting the sample with an antibody that binds FR ⁇ .
  • the antibody is selected from the group consisting of:
  • the antibody binds the same epitope as the MORAb-003 antibody.
  • the antibody includes SEQ ID NO: 1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • the antibody is the MOV18 antibody.
  • the antibody binds the same epitope as the MOV18 antibody.
  • the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21).
  • the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the level of FR ⁇ in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody.
  • the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • the level of FR ⁇ is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • control sample is a standardized control level of FR ⁇ in a healthy subject.
  • control sample is a sample previously obtained from the subject.
  • the sample is treated with guanidine prior to determining the level of FR ⁇ in the sample.
  • the sample is diluted prior to determining the level of FR ⁇ in the sample.
  • the sample is centrifuged, vortexed, or both, prior to determining the level of FR ⁇ in the sample.
  • the present invention provides a method for predicting whether a subject suffering from an FR ⁇ expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample derived from the subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • FR ⁇ folate receptor alpha
  • the present invention provides a method for predicting whether a subject suffering from an FR ⁇ expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the urine sample derived from the subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the urine sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • FR ⁇ folate receptor alpha
  • the present invention provides a method for predicting whether a subject suffering from an FR ⁇ expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the serum sample derived from the subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the serum sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • FR ⁇ folate receptor alpha
  • the FR ⁇ -expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer.
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing lung cancer is non-small cell lung cancer, such as adenocarcinoma.
  • the present invention provides methods for predicting whether a subject suffering from ovarian cancer will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FR ⁇ which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject will respond to treatment with MORAb-003.
  • FR ⁇ folate receptor alpha
  • the level of FR ⁇ is determined by contacting the sample with an antibody that binds FR ⁇ .
  • the antibody is selected from the group consisting of:
  • the antibody binds the same epitope as the MORAb-003 antibody.
  • the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • the antibody is the MOV 18 antibody.
  • the antibody binds the same epitope as the MOV18 antibody.
  • the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21).
  • the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the level of FR ⁇ in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody.
  • the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • the level of FR ⁇ is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • control sample is a standardized control level of FR ⁇ in a healthy subject.
  • the sample is treated with guanidine prior to determining the level of FR ⁇ in the sample.
  • the sample is diluted prior to determining the level of FR ⁇ in the sample.
  • the sample is centrifuged, vortexed, or both, prior to determining the level of FR ⁇ in the sample.
  • the present invention provides a method for predicting whether a subject suffering from an FR ⁇ expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample with the level of FR ⁇ in a control sample, wherein a difference between the levels of FR ⁇ in the sample derived from the subject and in the control sample is an indication that the subject will respond to treatment with MORAb-003; wherein the level of FR ⁇ in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid
  • the MORAb-003 for treatment is (a) an antibody that comprises the heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the light chain amino acid sequence as set forth in SEQ ID NO:8; (b) an antibody that binds the same epitope as the MORAb-003 antibody; or (c) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • SEQ ID NO:1 GTFSGYGLS
  • SEQ ID NO:2 MISGGSYTYYADSVKG
  • SEQ ID NO:3 HGDDPAWFAY
  • SEQ ID NO:4
  • the present invention provides methods of treating a subject having ovarian cancer or lung cancer by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a sample derived from said subject (for example, urine, serum, plasma or ascites); and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the sample derived from said subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with ovarian cancer or lung cancer; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • FR ⁇ folate receptor alpha
  • the present invention provides methods of treating a subject having ovarian cancer or lung cancer by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a urine sample derived from said subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the urine sample derived from said subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with ovarian cancer or lung cancer; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • FR ⁇ folate receptor alpha
  • the present invention provides methods of treating a subject having ovarian cancer or lung cancer by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a serum sample derived from said subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the serum sample derived from said subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with ovarian cancer or lung cancer; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • FR ⁇ folate receptor alpha
  • the present invention provides methods for treating a subject suffering from ovarian cancer by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FR ⁇ which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject will respond to treatment with MORAb-003; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • FR ⁇ folate receptor alpha
  • the level of FR ⁇ which is not bound to a cell in the sample derived from said subject is assessed by contacting the sample with an antibody that binds FR ⁇ .
  • the level of FR ⁇ is determined by contacting the sample with an antibody that binds FR ⁇ .
  • the antibody is selected from the group consisting of:
  • the antibody binds the same epitope as the MORAb-003 antibody.
  • the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • the antibody is the MOV 18 antibody.
  • the antibody binds the same epitope as the MOV18 antibody.
  • the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21).
  • the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the level of FR ⁇ in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody.
  • the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • the level of FR ⁇ is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • control sample is a standardized control level of FR ⁇ in a healthy subject.
  • the sample is treated with guanidine prior to determining the level of FR ⁇ in the sample.
  • the sample is diluted prior to determining the level of FR ⁇ in the sample.
  • the sample is centrifuged, vortexed, or both, prior to determining the level of FR ⁇ in the sample.
  • the present invention provides a method for treating a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample with the level of FR ⁇ in a control sample, wherein a difference between the levels of FR ⁇ in the sample derived from the subject and in the control sample is an indication that the subject will respond to treatment with MORAb-003; wherein the level of FR ⁇ in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobilized to
  • the MORAb-003 for treatment is (a) an antibody that comprises the heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the light chain amino acid sequence as set forth in SEQ ID NO:8; (b) an antibody that binds the same epitope as the MORAb-003 antibody; or (c) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • SEQ ID NO:1 GTFSGYGLS
  • SEQ ID NO:2 MISGGSYTYYADSVKG
  • SEQ ID NO:3 HGDDPAWFAY
  • SEQ ID NO:4
  • the present invention provides a kit for assessing whether a subject is afflicted with an FR ⁇ -expressing cancer or for assessing the progression of an FR ⁇ -expressing cancer in a subject, the kit including means for determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from the subject; and instructions for use of the kit to assess whether the subject is afflicted with an FR ⁇ -expressing cancer or to assess the progression of an FR ⁇ -expressing cancer.
  • FR ⁇ folate receptor alpha
  • the FR ⁇ -expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer.
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is non-small cell lung cancer, for example, adenocarcinoma.
  • the sample is either urine, serum, plasma or ascites.
  • the means includes a folate receptor alpha (FR ⁇ ) binding agent, for example, an antibody.
  • FR ⁇ folate receptor alpha
  • the antibody is selected from the group consisting of
  • the antibody is labeled including, but not limited to, a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • the kit includes a means for obtaining a sample from the subject.
  • the present invention is based, at least in part, on the unexpected discovery that folate receptor alpha (FR ⁇ ), not bound to a cell, is found at elevated levels in the body fluids, for example, urine or serum, of a subject having an FR ⁇ -expressing cancer such as lung or ovarian cancer as compared to a control sample. Moreover, the present invention is based, at least in part, on the identification of an immunological assay that exhibits the necessary sensitivity for assessing FR ⁇ levels in samples, where prior attempts to do so repeatedly failed. As a result, the present invention provides methods for diagnosing an FR ⁇ -expressing cancer by assessing levels of an FR ⁇ not bound to a cell in samples derived from the subject.
  • FR ⁇ folate receptor alpha
  • the present invention overcomes the challenges observed during prior attempts to develop an FR ⁇ based diagnostic assay for FR ⁇ -expressing cancers such as ovarian cancer by providing an immunological assay capable of accurately assessing levels of FR ⁇ not bound to a cell in samples.
  • kits for assessing whether a subject has or is at risk for developing an FR ⁇ -expressing cancer and, further, for assessing the progression of FR ⁇ -expressing cancer are provided.
  • the methods involve the comparison of levels of FR ⁇ not bound to a cell in samples, for example, urine and serum, as compared to control levels, in assessing the presence, degree or risk of development of ovarian cancer in the subject.
  • the methods involve the use of the MORAb-003 antibody, antibodies that bind the same epitope as the MORAb-003 antibody or antibodies having SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3, in assessing the levels of FR ⁇ not bound to a cell in a sample, e.g., urine or serum.
  • a sample e.g., urine or serum.
  • the MOV18 antibody or an antibody that binds the same epitope of the MOV18 antibody, the 548908 antibody, an antibody that binds the same epitope of the 548908 antibody, the 6D398 antibody or an antibody that binds the same epitope of the 548908 antibody may be used in accordance with the methods of the present invention.
  • an element means one element or more than one element.
  • the term "subject” refer to human and non-human animals, including veterinary subjects.
  • the term “non-human animal” includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, mice, rabbits, sheep, dog, cat, horse, cow, chickens, amphibians, and reptiles.
  • the subject is a human.
  • cancer or "tumor” are well known in the art and refer to the presence, e.g., in a subject, of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Cancer cells are often in the form of a tumor, but such cells may exist alone within a subject, or may be non-tumorigenic cancer cells, such as leukemia cells. As used herein, the term "cancer” includes pre-malignant as well as malignant cancers.
  • an "FR ⁇ -expressing cancer” includes any type of cancer characterized in that the cancer cells express FR ⁇ .
  • the FR ⁇ expressing cancer includes cancerous conditions characterized in that the cancer cells are capable of secreting, shedding, exporting or releasing FR ⁇ in such a manner that elevated levels of FR ⁇ are detectable in a biological sample from the subject.
  • FR ⁇ -expressing cancers include, but are not limited to, lung cancer (e.g., bronchioalveolar carcinomas, carcinoid tumors, and non-small cell lung cancers, such as adenocarcinomas); mesothelioma; ovarian cancer; renal cancer; brain cancer (e.g., anaplastic ependymoma and cerebellar juvenile pilocytic astrocytoma); cervical cancer; nasopharyngeal cancer; mesodermally derived tumor; squamous cell carcinoma of the head and neck; endometrial cancer; endometrioid adenocarcinomas of the ovary, serous cystadenocarcinomas, breast cancer; bladder cancer; pancreatic cancer; bone cancer (e.g., high-grade osteosarcoma); pituitary cancer (e.g., pituitary adenoma).
  • lung cancer e.g., bronchioalveolar carcinomas, carcino
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is lung cancer such as non-small cell lung cancer.
  • the FR ⁇ -expressing cancer is colorectal cancer and medullary thyroid cancer.
  • a subject who is "afflicted with an FR ⁇ -expressing cancer” is one who is clinically diagnosed with such a cancer by a qualified clinician (for example, by the methods of the present invention), or one who exhibits one or more signs or symptoms (for example, elevated levels of FR ⁇ in biological fluids) of such a cancer and is subsequently clinically diagnosed with such a cancer by a qualified clinician (for example, by the methods of the present invention).
  • a non-human subject that serves as an animal model of FR ⁇ -expressing cancer may also fall within the scope of the term a subject "afflicted with an FR ⁇ -expressing cancer.”
  • ovarian cancer refers to the art recognized disease and includes each of epithelial ovarian cancer (EOC; >90% of ovarian cancer in Western countries), germ cell tumors ( circa 2-3% of ovarian cancer), and stromal ovarian cancer. Ovarian cancer is stratified into different groups based on the differentiation of the tumor tissue. In grade I, the tumor tissue is well differentiated. In grade II, tumor tissue is moderately well differentiated. In grade III, the tumor tissue is poorly differentiated. This grade correlates with a less favorable prognosis than grades I and II.
  • EOC epithelial ovarian cancer
  • Stage I is generally confined within the capsule surrounding one (stage IA) or both (stage IB) ovaries, although in some stage I (i.e. stage IC) cancers, malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries.
  • Stage II involves extension or metastasis of the tumor from one or both ovaries to other pelvic structures.
  • stage IIA the tumor extends or has metastasized to the uterus, the fallopian tubes, or both.
  • Stage IIB involves extension of the tumor to the pelvis.
  • Stage IIC is stage IIA or IIB in which malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries.
  • the tumor comprises at least one malignant extension to the small bowel or the omentum, has formed extrapelvic peritoneal implants of microscopic (stage IIIA) or macroscopic ( ⁇ 2 centimeter diameter, stage IIIB; > 2 centimeter diameter, stage IIIC) size, or has metastasized to a retroperitoneal or inguinal lymph node (an alternate indicator of stage IIIC).
  • stage IV distant (i.e. non-peritoneal) metastases of the tumor can be detected.
  • ovarian cancer Each of the foregoing types, groups and stages of ovarian cancer are encompassed by the term "ovarian cancer" as used herein.
  • lung cancer refers to a disease in tissues of the lung involving uncontrolled cell growth, which, in some cases, leads to metastasis. Lung cancer is the most common cause of cancer-related death in men and women. The majority of primary lung cancers are carcinomas of the lung, derived from epithelial cells. The main types of lung cancer are small cell lung carcinoma (SCLC) and non-small cell lung carcinoma (NSCLC). In a particular embodiment, the FR ⁇ -expressing cancer is a non-small cell lung cancer.
  • SCLC small cell lung carcinoma
  • NSCLC non-small cell lung carcinoma
  • the FR ⁇ -expressing cancer is a non-small cell lung cancer.
  • SCLC small cell lung carcinoma
  • non-small cell lung cancer also known as non-small cell lung carcinoma (NSCLC) refers to epithelial lung cancer other than small cell lung carcinoma (SCLC).
  • SCLC small cell lung carcinoma
  • adenocarcinoma adenocarcinoma
  • squamous cell lung carcinoma adenocarcinoma
  • large cell lung carcinoma adenocarcinoma
  • Other less common types of non-small cell lung cancer include pleomorphic, carcinoid tumor, salivary gland carcinoma, and unclassified carcinoma.
  • Adenocarcinomas account for approximately 40% of lung cancers, and are the most common type of lung cancer in people who have never smoked.
  • Squamous cell carcinomas account for about 25% of lung cancers.
  • SCLC is categorized as limited disease confined to the ipsilateral hemithorax or as extensive disease with metastasis beyond the ipsilateral hemithorax.
  • NSCLC may be categorized using the tumor-nodes-metastasis (TNM) staging system.
  • TNM tumor-nodes-metastasis
  • Spira Spira J & Ettinger, D.S. Multidisciplinary management of lung cancer, N Engl J Med, 350:382- (2004 ) (hereinafter Spira); Greene FL, Page DL, Fleming ID, Fritz AG, Balch CM, Haller DG, et al (eds).
  • NSCLC is typically treated according to the stage of cancer determined by the following classification scheme (see http://www.cancer.gov/cancertopics/pdq/treatment/non-small-cell-lung/Patient/page2#Keypoint10).
  • stage 0 carcinoma in situ
  • abnormal cells are found in the lining of the airways. These abnormal cells may become cancer and spread into nearby normal tissue. Stage 0 is also called carcinoma in situ.
  • Stage I in which cancer has formed, is divided into stages IA and IB.
  • the tumor is in the lung only and is 3 centimeters or smaller.
  • the cancer has not spread to the lymph nodes and one or more of the following is true: (i) The tumor is larger than 3 centimeters but not larger than 5 centimeters; (ii) cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus; (iii) cancer has spread to the innermost layer of the membrane that covers the lung; (iv) part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus.
  • Stage IIA cancer has spread to certain lymph nodes on the same side of the chest as the primary tumor; the cancer is (a) 5 cm or smaller, (b) has spread to the main bronchus, and/or (c) has spread to the innermost layer of the lung lining.
  • cancer has not spread to lymph nodes; the cancer is (d) larger than 5 cm but not larger than 7 cm, (e) has spread to the main bronchus, and/or (f) has spread to the innermost layer of the lung lining.
  • Part of the lung may have collapsed or become inflamed.
  • Stage IIA is divided into two sections depending on the size of the tumor, where the tumor is found, and whether there is cancer in the lymph nodes.
  • cancer has spread to lymph nodes on the same side of the chest as the tumor.
  • the lymph nodes with cancer are within the lung or near the bronchus.
  • the tumor is not larger than 5 centimeters
  • cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus
  • cancer has spread to the innermost layer of the membrane that covers the lung
  • part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus.
  • cancer has not spread to lymph nodes and one or more of the following is true: (i) the tumor is larger than 5 centimeters but not larger than 7 centimeters, (ii) cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus, (iii) cancer has spread to the innermost layer of the membrane that covers the lung, (iv) part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus.
  • cancer has spread to certain lymph nodes on the same side of the chest as the primary tumor; the cancer is (a) larger than 5 cm but not larger than 7 cm, (b) has spread to the main bronchus, and/or (c) has spread to the innermost layer of the lung lining. Part of the lung may have collapsed or become inflamed.
  • the cancer is larger than 7 cm; (e) has spread to the main bronchus, (f) the diaphragm, (g) the chest wall or the lining of the chest wall; and/or (h) has spread to the membrane around the heart.
  • Stage IIB is divided into two sections depending on the size of the tumor, where the tumor is found, and whether there is cancer in the lymph nodes. In the first section, cancer has spread to nearby lymph nodes on the same side of the chest as the tumor. The lymph nodes with cancer are within the lung or near the bronchus.
  • the tumor is larger than 5 centimeters but not larger than 7 centimeters
  • cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus
  • cancer has spread to the innermost layer of the membrane that covers the lung
  • part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus.
  • cancer has not spread to lymph nodes and one or more of the following is true: (i) the tumor is larger than 7 centimeters, (ii) cancer has spread to the main bronchus (and is less than 2 centimeters below where the trachea joins the bronchus), the chest wall, the diaphragm, or the nerve that controls the diaphragm, (iii) cancer has spread to the membrane around the heart or lining the chest wall, (iv) the whole lung has collapsed or developed pneumonitis (inflammation of the lung), (v) there are one or more separate tumors in the same lobe of the lung.
  • Stage IIIA is divided into three sections depending on the size of the tumor, where the tumor is found, and which lymph nodes have cancer (if any).
  • cancer has spread to lymph nodes on the same side of the chest as the tumor.
  • the lymph nodes with cancer are near the sternum (chest bone) or where the bronchus enters the lung.
  • the tumor may be any size; part of the lung (where the trachea joins the bronchus) or the whole lung may have collapsed or developed pneumonitis (inflammation of the lung); there may be one or more separate tumors in the same lobe of the lung; and cancer may have spread to any of the following: (i) main bronchus, but not the area where the trachea joins the bronchus, (ii) chest wall, (iii) diaphragm and the nerve that controls it, (iv) membrane around the lung or lining the chest wall, (iv) membrane around the heart. In the second section of Stage IIIA, cancer has spread to lymph nodes on the same side of the chest as the tumor.
  • the lymph nodes with cancer are within the lung or near the bronchus.
  • the tumor may be any size; the whole lung may have collapsed or developed pneumonitis (inflammation of the lung); there may be one or more separate tumors in any of the lobes of the lung with cancer; and cancer may have spread to any of the following: (i) main bronchus, but not the area where the trachea joins the bronchus, (ii) chest wall, (iii) diaphragm and the nerve that controls it, (iv) membrane around the lung or lining the chest wall, (v) heart or the membrane around it, (vi) major blood vessels that lead to or from the heart, (vi) trachea, (vii) esophagus, (viii) nerve that controls the larynx (voice box), (ix) sternum (chest bone) or backbone, (x) carina (where the trachea joins the bronchi).
  • cancer has not spread to the lymph nodes and the tumor may be any size, and cancer has spread to any of the following: (i) heart, (ii) major blood vessels that lead to or from the heart, (iii) trachea, (iv) esophagus, (v) nerve that controls the larynx (voice box), (vi) sternum (chest bone) or backbone, (vi) carina (where the trachea joins the bronchi).
  • Stage IIIB is divided into two sections depending on the size of the tumor, where the tumor is found, and which lymph nodes have cancer.
  • cancer has spread to lymph nodes above the collarbone or to lymph nodes on the opposite side of the chest as the tumor; the tumor may be any size; part of the lung (where the trachea joins the bronchus) or the whole lung may have collapsed or developed pneumonitis (inflammation of the lung); there may be one or more separate tumors in any of the lobes of the lung with cancer; and cancer may have spread to any of the following: (i) main bronchus, (ii) chest wall, (iii) diaphragm and the nerve that controls it, (iv) membrane around the lung or lining the chest wall, (iv) heart or the membrane around it, (v) major blood vessels that lead to or from the heart, (vi) trachea, (vii) esophagus, (viii) nerve that controls the larynx (voice box), (ix)
  • cancer has spread to lymph nodes on the same side of the chest as the tumor; the lymph nodes with cancer are near the sternum (chest bone) or where the bronchus enters the lung; the tumor may be any size; there may be separate tumors in different lobes of the same lung; and cancer has spread to any of the following: (i) heart, (ii) major blood vessels that lead to or from the heart, (iii) trachea, (iv) esophagus, (v) nerve that controls the larynx (voice box), (vi) sternum (chest bone) or backbone, (vii) carina (where the trachea joins the bronchi).
  • the tumor may be any size and cancer may have spread to lymph nodes.
  • cancer may have spread to lymph nodes.
  • the lung cancer may be stratified into any of the preceding stages (e.g., occult, stage 0, stage IA, stage IB, stage IIA, stage IIB, stage IIIA, stage IIIB or stage IV) based on assessing of the levels of FR ⁇ not bound to a cell, such as a normal or cancerous cell, in a sample (for example, urine or serum) of a subject.
  • a cell such as a normal or cancerous cell
  • a sample for example, urine or serum
  • FR ⁇ alpha isoform of the high affinity receptor for folate.
  • Membrane bound FR ⁇ is attached to the cell surface by a glycosyl phosphatidylinositol (GPI) anchor), recycles between extracellular and endocytic compartments and is capable of transporting folate into the cell.
  • GPI glycosyl phosphatidylinositol
  • FR ⁇ is expressed in a variety of epithelial tissues including those of the female reproductive tract, placenta, breast, kidney proximal tubules, choroid plexus, lung and salivary glands. Soluble forms of FR ⁇ may be derived by the action of proteases or phospholipase on membrane anchored folate receptors.
  • the consensus nucleotide and amino acid sequences for human FR ⁇ are set forth herein as SEQ ID NOs: 24 and 25, respectively.
  • Variants for example, naturally occurring allelic variants or sequences containing at least one amino acid substitution, are encompassed by the terms as used herein.
  • the term "not bound to a cell” refers to FR ⁇ that is not attached to the cellular membrane of a cell, such as a cancerous cell.
  • the FR ⁇ not bound to a cell is unbound to any cell and is freely floating or solubilized in biological fluids, e.g. , urine or serum.
  • biological fluids e.g. , urine or serum.
  • the FR ⁇ may be shed, secreted or exported from normal or cancerous cells, for example, from the surface of cancerous cells, into biological fluids.
  • the "level" of folate receptor alpha not bound to a cell refers to the level of folate receptor alpha protein as determined using any method known in the art for the measurement of protein levels. Such methods include, for example, electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, fluid or gel precipitin reactions, absorption spectroscopy, a colorimetric assays, spectrophotometric assays, flow cytometry, immunodiffusion (single or double), solution phase assay, immunoelectrophoresis, Western blotting, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, and electrochemiluminescence immunoassay (exemplified below), and the like. In a preferred embodiment, the level is determined using antibody-based techniques, as described in more detail herein.
  • Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody.
  • Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • protein isolated from a subject sample e.g., urine or serum
  • a polyacrylamide gel electrophoresis and immobilized onto a solid phase support such as nitrocellulose.
  • the support can then be washed with suitable buffers followed by treatment with the labeled antibody.
  • the solid phase support can then be washed with the buffer a second time to remove unbound antibody.
  • the amount of bound label on the solid support can then be detected by conventional means.
  • Means of detecting proteins using electrophoretic techniques are well known to those of skill in the art (see generally, R. Scopes (1982) Protein Purification, Springer-Verlag, N.Y .; Deutscher, (1990) Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc., N.Y .).
  • Antibodies used in immunoassays to determine the level of expression of folate receptor alpha may be labeled with a detectable label.
  • the term "labeled", with regard to the binding agent or antibody, is intended to encompass direct labeling of the binding agent or antibody by coupling ( i.e., physically linking) a detectable substance to the binding agent or antibody, as well as indirect labeling of the binding agent or antibody by reactivity with another reagent that is directly labeled.
  • An example of indirect labeling includes detection of a primary antibody using a fluorescently labeled secondary antibody.
  • the antibody is labeled, e.g. , radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled.
  • the antibody is an antibody derivative (e.g ., an antibody conjugated with a substrate or with the protein or ligand of a protein-ligand pair (e.g. , biotin-streptavidin), or an antibody fragment (e.g. , a single-chain antibody, an isolated antibody hypervariable domain) which binds specifically with FR ⁇ not bound to a cell.
  • a protein-ligand pair e.g. , biotin-streptavidin
  • an antibody fragment e.g. , a single-chain antibody, an isolated antibody hypervariable domain
  • proteomic methods e.g. , mass spectrometry
  • Mass spectrometry is an analytical technique that consists of ionizing chemical compounds to generate charged molecules (or fragments thereof) and measuring their mass-to-charge ratios.
  • a sample is obtained from a subject, loaded onto the mass spectrometry, and its components (e.g., FR ⁇ ) are ionized by different methods (e.g., by impacting them with an electron beam), resulting in the formation of charged particles (ions).
  • the mass-to-charge ratio of the particles is then calculated from the motion of the ions as they transit through electromagnetic fields.
  • MALDI-TOF MS matrix-associated laser desorption/ionization time-of-flight mass spectrometry
  • SELDI-TOF MS surface-enhanced laser desorption/ionization time-of-flight mass spectrometry
  • in vivo techniques for determination of the level of FR ⁇ not bound to a cell include introducing into a subject a labeled antibody directed against FR ⁇ , which binds to and transforms FR ⁇ into a detectable molecule.
  • the presence, level, or location of the detectable FR ⁇ not bound to a cell in a subject may be determined using standard imaging techniques.
  • sample refers to a collection of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject.
  • the sample is a biological fluid containing FR ⁇ not bound to a cancerous cell.
  • Biological fluids are typically liquids at physiological temperatures and may include naturally occurring fluids present in, withdrawn from, expressed or otherwise extracted from a subject or biological source. Certain biological fluids derive from particular tissues, organs or localized regions and certain other biological fluids may be more globally or systemically situated in a subject or biological source.
  • biological fluids include blood, serum and serosal fluids, plasma, lymph, urine, cerebrospinal fluid, saliva, ocular fluids, cystic fluid, tear drops, feces, sputum, mucosal secretions of the secretory tissues and organs, vaginal secretions, gynecological fluids, ascites fluids such as those associated with non-solid tumors, fluids of the pleural, pericardial, peritoneal, abdominal and other body cavities, fluids collected by bronchial lavage and the like.
  • the sample is urine or serum.
  • the sample does not include ascites or is not an ascite sample.
  • the sample does not include peritoneal fluid or is not peritoneal fluid.
  • the sample is removed from the subject. In another embodiment, the sample is present within the subject.
  • Biological fluids may also include liquid solutions contacted with a subject or biological source, for example, cell and organ culture medium including cell or organ conditioned medium, lavage fluids and the like.
  • sample may be pre-treated by physical or chemical means prior to the assay.
  • samples for example, urine samples, were subjected to centrifugation, dilution and/or treatment with a solubilizing substance (e.g., guanidine treatment) prior to assaying the samples for FR ⁇ not bound to a cell.
  • solubilizing substance e.g., guanidine treatment
  • control sample refers to any clinically relevant control sample, including, for example, a sample from a healthy subject not afflicted with ovarian cancer, a sample from a subject having a less severe or slower progressing ovarian cancer than the subject to be assessed, a sample from a subject having some other type of cancer or disease, and the like.
  • a control sample may include a sample derived from one or more subjects.
  • a control sample may also be a sample made at an earlier timepoint from the subject to be assessed.
  • the control sample could be a sample taken from the subject to be assessed before the onset of the FR ⁇ expressing cancer such as lung or ovarian cancer, at an earlier stage of disease, or before the administration of treatment or of a portion of treatment.
  • the control sample may also be a sample from an animal model, or from a tissue or cell lines derived from the animal model, of the FR ⁇ expressing cancer such as lung or ovarian cancer.
  • the level of FR ⁇ not bound to a cell in a control sample that consists of a group of measurements may be determined based on any appropriate statistical measure, such as, for example, measures of central tendency including average, median, or modal values.
  • control level refers to an accepted or pre-determined level of FR ⁇ which is used to compare with the level of FR ⁇ in a sample derived from a subject.
  • control level of FR ⁇ is based on the level of FR ⁇ not bound to a cell in sample(s) from a subject(s) having slow disease progression.
  • control level of FR ⁇ not bound to a cell is based on the level in a sample from a subject(s) having rapid disease progression.
  • control level of FR ⁇ is based on the level of FR ⁇ not bound to a cell in a sample(s) from an unaffected, i.e., non-diseased, subject(s), i.e., a subject who does not have an FR ⁇ expressing cancer such as lung or ovarian cancer.
  • control level of FR ⁇ is based on the level of FR ⁇ not bound to a cell in a sample from a subject(s) prior to the administration of a therapy for ovarian cancer.
  • control level of FR ⁇ is based on the level of FR ⁇ not bound to a cell in a sample(s) from a subject(s) having an FR ⁇ expressing cancer such as lung or ovarian cancer that is not contacted with a test compound. In another embodiment, the control level of FR ⁇ is based on the level of FR ⁇ not bound to a cell in a sample(s) from a subject(s) not having an FR ⁇ expressing cancer such as lung or ovarian cancer that is contacted with a test compound.
  • control level of FR ⁇ is based on the level of FR ⁇ not bound to a cell in a sample(s) from an animal model of an FR ⁇ expressing cancer such as lung or ovarian cancer, a cell, or a cell line derived from the animal model of an FR ⁇ expressing cancer such as lung or ovarian cancer.
  • control is a standardized control, such as, for example, a control which is predetermined using an average of the levels of FR ⁇ not bound to a cell from a population of subjects having no FR ⁇ expressing cancer such as lung or ovarian cancer.
  • a control level of FR ⁇ is based on the level of FR ⁇ not bound to a cell in a non-cancerous sample(s) derived from the subject having an FR ⁇ expressing cancer such as lung or ovarian cancer.
  • the control level of FR ⁇ may be determined using the non-affected portion of the ovaries, and this control level may be compared with the level of FR ⁇ in an affected portion of the ovaries.
  • the control level of FR ⁇ may be determined using the non-affected portion of the lungs, and this control level may be compared with the level of FR ⁇ in an affected portion of the lungs.
  • a difference between the level of folate receptor alpha not bound to a cell in a sample from a subject (i.e., a test sample) and the level of folate receptor alpha not bound to a cell in a control sample refers broadly to any clinically relevant and/or statistically significant difference in the level of folate receptor alpha in the two samples.
  • the difference is selected based on a cutoff value determined using a receiver operating characteristic (ROC) analysis, an example of which is presented in Example 6.
  • ROC receiver operating characteristic
  • the difference must be greater than the limits of detection of the method for determining the level of FR ⁇ not bound to a cell. It is preferred that the difference be at least greater than the standard error of the assessment method, and preferably a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method. The difference may be assessed by any appropriate comparison, including any appropriate parametric or nonparametric descriptive statistic or comparison.
  • an increase in the level of FR ⁇ not bound to a cell may refer to a level in a test sample that is about two, and more preferably about three, about four, about five, about six, about seven, about eight, about nine, about ten or more times more than the level of FR ⁇ in the control sample.
  • An increase may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations above the average level of FR ⁇ in the control sample.
  • a decrease in the level of FR ⁇ not bound to a cell may refer to a level in a test sample that is preferably at least about two, and more preferably about three, about four, about five, about six, about seven, about eight, about nine, about ten or more times less than the level of FR ⁇ in the control sample.
  • a decrease may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations below the average level of FR ⁇ in the control sample.
  • the term "contacting the sample" with an FR ⁇ binding agent includes exposing the sample, or any portion thereof with the agent or antibody, such that at least a portion of the sample comes into contact with the agent or antibody.
  • the sample or portion thereof may be altered in some way, such as by subjecting it to physical or chemical treatments (e.g., dilution or guanidine treatment), prior to the act of contacting it with the agent or antibody.
  • antibody as used herein, comprises four polypeptide chains, two heavy (H) chains and two light (L) chains, interconnected by disulfide bonds, as well as any functional (i.e., antigen-binding) fragment, mutant, variant, or derivation thereof, which retains the essential epitope binding features of an Ig molecule.
  • mutant, variant, or derivative antibody formats are known in the art, and include molecules such as Fab fragments, Fab' fragments, F(ab') 2 fragments, Fd fragments, Fabc fragments, Sc antibodies (single chain antibodies), diabodies, individual antibody light chains, individual antibody heavy chains, chimeric fusions between antibody chains and the like.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass.
  • type e.g., IgG, IgE, IgM, IgD, IgA and IgY
  • class e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2 or subclass.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDRs complementarity determining regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively.
  • variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g. , effector cells) and the first component (Clq) of the classical complement system.
  • antigen-binding portion of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., FR ⁇ not bound to a cell). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the V L , V H , CL and CH1 domains; (ii) a F(ab') 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the V H and CH1 domains; (iv) a Fv fragment consisting of the V L and V H domains of a single arm of an antibody, (v) a dAb including VH and VL domains; (vi) a dAb fragment ( Ward et al.
  • V H domain a dAb which consists of a VH or a VL domain
  • CDR an isolated complementarity determining region
  • ix a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker.
  • the two domains of the Fv fragment, V L and V H are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the V L and V H regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al.
  • scFv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
  • antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
  • Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins.
  • antibody includes polyclonal antibodies, monoclonal antibodies, murine antibodies, chimeric antibodies, humanized antibodies, and human antibodies, and those that occur naturally or are recombinantly produced according to methods well known in the art.
  • an antibody for use in the methods of the invention is a bispecific antibody.
  • a "bispecific antibody” is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, (1990) Clin. Exp. Immunol. 79, 315-321 ; Kostelny et al. (1992) J. Immunol. 148, 1547-1553 .
  • an antibody for use in the methods of the invention is a camelid antibody as described in, for example, PCT Publication WO 94/04678 , the entire contents of which are incorporated herein by reference.
  • a region of the camelid antibody that is the small, single variable domain identified as V HH can be obtained by genetic engineering to yield a small protein having high affinity for a target, resulting in a low molecular weight, antibody-derived protein known as a "camelid nanobody”.
  • V HH camelid antibody
  • U.S. Pat. No. 5,759,808 see also Stijlemans et al., 2004 J. Biol. Chem. 279: 1256-1261 ; Dumoulin et al., 2003 Nature 424: 783-788 ; Pleschberger et al., 2003 Bioconjugate Chem. 14: 440-448 ; Cortez-Retamozo et al., 2002 Int. J.
  • a feature of the present invention is a camelid nanobody having high affinity for FR ⁇ .
  • an antibody for use in the methods of the invention is a diabody, a single chain diabody, or a di-diabody.
  • Diabodies are bivalent, bispecific molecules in which V H and V L domains are expressed on a single polypeptide chain, connected by a linker that is too short to allow for pairing between the two domains on the same chain.
  • the V H and V L domains pair with complementary domains of another chain, thereby creating two antigen binding sites (see e.g., Holliger et al., 1993 Proc. Natl. Acad. Sci. USA 90:6444-6448 ; Poljak et al., 1994 Structure 2:1121-1123 ).
  • Diabodies can be produced by expressing two polypeptide chains with either the structure V HA -V LB and V HB -V LA (V H -V L configuration), or V LA -V HB and V LB -V HA (V L -V H configuration) within the same cell. Most of them can be expressed in soluble form in bacteria.
  • Single chain diabodies are produced by connecting the two diabody-forming polypeptide chains with linker of approximately 15 amino acid residues (see Holliger and Winter, 1997 Cancer Immunol. Immunother., 45(3-4):128-30 ; Wu et al., 1996 Immunotechnology, 2(1):21-36 ).
  • scDb can be expressed in bacteria in soluble, active monomeric form (see Holliger and Winter, 1997 Cancer Immunol.
  • a diabody can be fused to Fc to generate a "di-diabody" (see Lu et al., 2004 J. Biol. Chem., 279(4):2856-65 ).
  • FR ⁇ binding molecules that exhibit functional properties of antibodies but derive their framework and antigen binding portions from other polypeptides (e.g., polypeptides other than those encoded by antibody genes or generated by the recombination of antibody genes in vivo ) may also be used in the methods of the present invention.
  • the antigen binding domains e.g ., FR ⁇ binding domains
  • the antigen binding domains of these binding molecules are generated through a directed evolution process. See U.S. Pat. No. 7,115,396 .
  • Molecules that have an overall fold similar to that of a variable domain of an antibody are appropriate scaffold proteins.
  • Scaffold proteins suitable for deriving antigen binding molecules include fibronectin or a fibronectin dimer, tenascin, N-cadherin, E-cadherin, ICAM, titin, GCSF-receptor, cytokine receptor, glycosidase inhibitor, antibiotic chromoprotein, myelin membrane adhesion molecule P0, CD8, CD4, CD2, class I MHC, T-cell antigen receptor, CD1, C2 and I-set domains of VCAM-1, I-set immunoglobulin domain of myosin-binding protein C, I-set immunoglobulin domain of myosin-binding protein H, I-set immunoglobulin domain of telokin, NCAM, twitchin, neuroglian, growth hormone receptor, erythropoietin receptor, prolactin receptor, interferon-gamma receptor, P-galactosidase/glucuronidase, ⁇ -glucuronidase, transglutamina
  • Specific binding when used in the context of antibodies, or antibody fragments, represents binding via domains encoded by immunoglobulin genes or fragments of immunoglobulin genes to one or more epitopes of a protein of interest, but which do not substantially recognize and bind other molecules in a sample containing a mixed population of antigenic molecules.
  • an antibody binds to a cognate antigen with a Kd of less than about 1x10 -8 M, as measured by a surface plasmon resonance assay or a cell binding assay.
  • a folate receptor alpha "binding agent” includes an antibody that binds FR ⁇ not bound to a cell as well as non-antibody binding agents.
  • a library of clones can be created in which sequences in regions of the scaffold protein that form antigen binding surfaces (e.g., regions analogous in position and structure to CDRs of an antibody variable domain immunoglobulin fold) are randomized.
  • Library clones are tested for specific binding to the antigen of interest (e.g., FR ⁇ ) and for other functions (e.g., inhibition of biological activity of FR ⁇ ). Selected clones can be used as the basis for further randomization and selection to produce derivatives of higher affinity for the antigen.
  • High affinity binding molecules are generated, for example, using the tenth module of fibronectin III ( 10 Fn3) as the scaffold, described in U.S. Pat. Nos. 6,818,418 and 7,115,396 ; Roberts and Szostak, 1997 Proc. Natl. Acad. Sci USA 94:12297 ; U.S. Pat. No. 6,261,804 ; U.S. Pat. No. 6,258,558 ; and Szostak et al. WO98/31700 , the entire contents of each of which are incorporated herein by reference.
  • Non-antibody binding molecules can be produced as dimers or multimers to increase avidity for the target antigen.
  • the antigen binding domain is expressed as a fusion with a constant region (Fc) of an antibody that forms Fc-Fc dimers. See, e.g., U.S. Pat. No. 7,115,396 , the entire contents of which are incorporated herein by reference.
  • an “antigen” is a molecule recognized by the immune system; the term originally came from “antibody generator” and includes a molecule that binds specifically to an antibody.
  • an antigen is characterized by its ability to be bound at the antigen-binding site of an antibody.
  • the antigen is FR ⁇ , such as FR ⁇ which is not bound to a cell FR ⁇ or a portion thereof.
  • epitope refers to the molecular surface features of an antigen, e.g., FR ⁇ , capable of being bound by an antibody.
  • Antigenic molecules normally being "large” biological polymers, usually present several surface features that can act as points of interaction for specific antibodies. Any such distinct molecular feature constitutes an epitope. Most antigens therefore have the potential to be bound by several distinct antibodies, each of which is typically specific to a particular epitope.
  • a binding agent e.g., antibody, binds to an epitope on FR ⁇ which is available in the form of the receptor which is not bound to a cell but not in the membrane bound form of the receptor.
  • the antibody may bind to the same epitope on FR ⁇ to which MORAB-003 binds.
  • progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer includes the progression of such a cancer from a less severe to a more severe state. This could include an increase in the number or severity of tumors, the degree of metastasis, the speed with which the cancer is growing and spreading, and the like.
  • progression of ovarian cancer includes the progression of such a cancer from a less severe to a more severe state, such as the progression from Stage I to Stage II, from Stage II to Stage III, etc.
  • the phrase "progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer” may refer to the regression of an FR ⁇ -expressing cancer from a more severe state to a less severe state.
  • “the progression of ovarian cancer” refers to the regression from Stage IV to Stage III, from Stage III to Stage II, etc.
  • the "progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer” may refer to the survival rate determined from the beginning of symptoms of the FR ⁇ -expressing cancer, or to the survival rate from the time of diagnosis of the FR ⁇ -expressing cancer.
  • stratifying refers to characterizing an FR ⁇ expressing cancer, for example, ovarian or lung cancer, into an appropriate stage based, for example, on the degree of the spread of the cancer, as well accepted stratifications in the art.
  • stratifying includes characterizing the FR ⁇ expressing cancer into Stage I, Stage II, Stage III or Stage IV.
  • Stage I refers to cancers that are localized to one part of the body.
  • Stages II and III refer to cancers that are locally advanced, wherein a distinction between the stages are often specific to the particular cancer.
  • Stage IV refers to cancers that have often metastasized, or spread to other organs or throughout the body.
  • the term "survival” refers to the continuation of life of a subject which has been treated for cancer.
  • survival refers to the failure of a tumor to recur.
  • the term "recur” refers to the re-growth of tumor or cancerous cells in a subject in whom primary treatment for the tumor has been administered.
  • the tumor may recur in the original site or in another part of the body.
  • a tumor that recurs is of the same type as the original tumor for which the subject was treated. For example, if a subject had an ovarian cancer tumor, was treated and subsequently developed another ovarian cancer tumor, the tumor has recurred.
  • a cancer can recur in a different organ or tissue than the one where it originally occurred.
  • the present invention is based, at least in part, on the unexpected discovery that folate receptor alpha (FR ⁇ ), not bound to a cell, is found at elevated levels in the body fluids, for example, urine or serum, of a subject having an FR ⁇ -expressing cancer as compared to a control sample. Moreover, the present invention is based, at least in part, on the identification of an immunological assay that exhibits the necessary sensitivity for assessing levels of FR ⁇ not bound to a cells in samples, where prior attempts to do so had repeatedly failed.
  • FR ⁇ folate receptor alpha
  • the present invention overcomes the challenges observed during prior attempts to develop an FR ⁇ -based diagnostic assay for FR ⁇ -expressing cancer such as lung or ovarian cancer by providing an immunological assay capable of accurately assessing levels of FR ⁇ not bound to a cell in a sample, e.g., urine or serum.
  • kits for assessing whether a subject has or is at risk for developing an FR ⁇ -expressing cancer and, further, for assessing the progression of an FR ⁇ -expressing cancer are provided.
  • the methods involve the comparison of levels of FR ⁇ not bound to a cell in samples, for example, urine and serum, as compared to control levels, in assessing the presence, degree or risk of development of ovarian cancer in the subject.
  • the present invention provides diagnostic methods for assessing whether a subject is afflicted with an FR ⁇ -expressing cancer, such as lung or ovarian cancer, prognostic methods for predicting the progression of an FR ⁇ -expressing cancer such as lung or ovarian cancer, and risk assessment methods for assessing the level of risk that a subject will develop the FR ⁇ -expressing cancer. Furthermore, the invention provides stratification methods for stratifying an FR ⁇ -expressing cancer such as lung or ovarian cancer subject into cancer therapy groups.
  • the various aspects and embodiments of the invention discussed here are intended to be non-limiting and to encompass all possible combinations of the specific embodiments mentioned, which may apply to any of the methods and kits discussed herein or claimed below.
  • the methods of the present invention can be practiced in conjunction with any other method used by the skilled practitioner to diagnose an FR ⁇ -expressing cancer, predict the progression of an FR ⁇ -expressing cancer, or to assess the level of risk that a subject will develop an FR ⁇ -expressing cancer.
  • the invention provides a method of assessing whether a subject is afflicted with an FR ⁇ -expressing cancer, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a sample (such as urine or serum) derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with an FR ⁇ -expressing cancer.
  • FR ⁇ folate receptor alpha
  • the level of FR ⁇ in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FR ⁇ not bound to a cell and is selected from the group consisting of (a) an antibody that binds the same epitope as the MORAb-003 antibody; and (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • the sample is selected from the group consisting of urine and serum.
  • the present invention provides a method of assessing whether a subject is afflicted with an FR ⁇ -expressing cancer such as lung cancer or ovarian cancer, the method comprising determining the level of folate receptor alpha (FR ⁇ ) not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) in the urine sample derived from the subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the urine sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with an FR ⁇ -expressing cancer.
  • an FR ⁇ -expressing cancer such as lung cancer or ovarian cancer
  • the invention provides a method of assessing whether a subject is afflicted with an FR ⁇ -expressing cancer, by determining the level of folate receptor alpha (FR ⁇ ) not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FR ⁇ ) in the serum sample derived from the subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the serum sample derived from the subject and the level of FR ⁇ in the control sample is an indication that the subject is afflicted with an FR ⁇ -expressing cancer.
  • the subject has not been treated with an agent, such as a steroid, that enhances the levels of FR ⁇ in serum.
  • the FR ⁇ -expressing cancer is ovarian cancer and the subject has not been treated with an agent, such as a steroid, that enhances the levels of FR ⁇ in serum.
  • FR ⁇ -expressing cancers include cancers characterized in that the cancer cells express FRa.
  • the FR ⁇ is released from the cancer cells, for example, from the surface of the cancer cell, and into the biological fluids of the subject.
  • FR ⁇ -expressing cancers include lung cancer (e.g ., bronchioalveolar carcinomas, carcinoid tumors, and non-small cell lung cancers, such as adenocarcinomas); mesothelioma; ovarian cancer; renal cancer; brain cancer (e.g ., anaplastic ependymoma and cerebellar juvenile pilocytic astrocytoma); cervical cancer; nasopharyngeal cancer; mesodermally derived tumor; squamous cell carcinoma of the head and neck; endometrial cancer; endometrioid adenocarcinomas of the ovary, serous cystadenocarcinomas, breast cancer; bladder cancer; pancreatic cancer; bone cancer (e.g., high-grade osteosarcoma); and pituitary cancer (e.g., pituitary adenoma).
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is lung cancer.
  • the lung cancer is non-small cell lung carcinoma (NSCLC).
  • NSCLC non-small cell lung carcinoma
  • the NSCLC is selected from the group consisting of adenocarcinoma, squamous cell lung carcinoma, large cell lung carcinoma, pleomorphic NSCLC, carcinoid tumor, salivary gland carcinoma, and unclassified carcinoma.
  • the NSCLC is adenocarcinoma.
  • the lung cancer is small cell lung carcinoma (SCLC).
  • SCLC small cell lung carcinoma
  • the lung cancer is bronchioalveolar carcinoma.
  • the lung cancer is a lung carcinoid tumor.
  • the present invention also provides methods to assess whether a subject is afflicted with ovarian cancer by determining the level of folate receptor alpha (FR ⁇ ) not bound to a cell in a urine sample derived from the subject, wherein the presence of FR ⁇ in the urine sample at a concentration of greater than about 3000 a.u./ml is an indication that the subject is afflicted with ovarian cancer.
  • FR ⁇ folate receptor alpha
  • the present invention provides a method of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FR ⁇ ) in a urine sample derived from the subject, wherein the presence of FR ⁇ in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer or wherein a concentration of less than about 9100 pg/ml is an indication that the subject is not afflicted with ovarian cancer.
  • FR ⁇ folate receptor alpha
  • the levels of FR ⁇ not bound to a cell in a sample are compared with the levels of FR ⁇ in a control sample, wherein a difference between the levels is an indication that the subject is afflicted with an FR ⁇ -expressing cancer such as lung or ovarian cancer.
  • the difference constitutes an increase in the level of FR ⁇ not bound to a cell in the sample derived from the subject as compared with the level of FR ⁇ in the control sample, wherein this increase is indicative of the presence or growth of FR ⁇ -expressing cancer.
  • the difference constitutes a decrease in the level of FR ⁇ , wherein the decrease is indicative of the absence or regression of FRa-expressing cancer.
  • a difference between the level of folate receptor alpha not bound to a cell in a sample from a subject (i.e., a test sample) and the level of folate receptor alpha in a control sample refers broadly to any clinically relevant change (including an increase or a decrease) and/or statistically significant difference in the level of folate receptor alpha in the two samples.
  • the difference is selected based on a cutoff value determined using a receiver operating characteristic (ROC) analysis, an example of which is presented in Example 6. The optimal cutoff value may vary depending on the assay methods and conditions employed.
  • ROC receiver operating characteristic
  • the difference must be greater than the limits of detection of the method for determining the level of FR ⁇ not bound to a cell. It is preferred that the difference be at least greater than the standard error of the assessment method, and preferably a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method.
  • the difference may be assessed by any appropriate comparison, including any appropriate parametric or nonparametric descriptive statistic or comparison. For example, "an increase" in the level of FR ⁇ may refer to a level that exceeds a cutoff value determined using an ROC analysis.
  • a level in a test sample that is two, and more preferably about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900% or about 1000% more than the level of FR ⁇ in the control sample.
  • An increase may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations above the average level of FR ⁇ in the control sample.
  • a decrease in the level of FR ⁇ not bound to a cell may refer to a level in a test sample that does not exceed a cutoff value determined using an ROC analysis. It may also refer to a level in a test sample that is about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, or about 90% less than the level of FR ⁇ in the control sample. A decrease may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations below the average level of FR ⁇ in the control sample.
  • Samples useful in the methods and kits of the invention include any tissue, cell, biopsy, or bodily fluid that may contain detectable levels of FR ⁇ not bound to a cell.
  • a sample may be a tissue, a cell, whole blood, plasma, buccal scrape, saliva, cerebrospinal fluid, stool, or bronchoalveolar lavage.
  • the sample is FR ⁇ -expressing tumor sample or a sample of tissues or cells where FR ⁇ -expressing cancer may be found.
  • the sample is a urine or serum sample.
  • Body samples may be obtained from a subject by a variety of techniques known in the art including, for example, by the use of a biopsy or by scraping or swabbing an area or by using a needle to aspirate bodily fluids. Methods for collecting various body samples are well known in the art.
  • Samples suitable for detecting and quantitating the FR ⁇ protein level may be fresh, frozen, or fixed according to methods known to one of skill in the art. Suitable tissue samples are preferably sectioned and placed on a microscope slide for further analyses. Solid samples, i.e., tissue samples, may be solubilized and/or homogenized and subsequently analyzed as soluble extracts. Liquid samples may also be subjected to physical or chemical treatments. In some embodiments, urine samples are treated by centrifugation, vortexing, dilution and/or treatment with a solubilizing substance (such as, for example, guanidine treatment).
  • a solubilizing substance such as, for example, guanidine treatment.
  • a freshly obtained biopsy sample is frozen using, for example, liquid nitrogen or difluorodichloromethane.
  • the frozen sample is mounted for sectioning using, for example, OCT, and serially sectioned in a cryostat.
  • the serial sections are collected on a glass microscope slide.
  • the slides may be coated with, for example, chrome-alum, gelatine or poly-L-lysine to ensure that the sections stick to the slides.
  • samples are fixed and embedded prior to sectioning.
  • a tissue sample may be fixed in, for example, formalin, serially dehydrated and embedded in, for example, paraffin.
  • any method known in the art to be suitable for detecting and quantitating FR ⁇ not bound to a cell may be used (either at the nucleic acid or, preferably, at the protein level), as described in section (B) below.
  • Exemplary methods are well known in the art and include but are not limited to western blots, northern blots, southern blots, immunohistochemistry, solution phase assay, ELISA, e.g ., amplified ELISA, immunoprecipitation, immunofluorescence, flow cytometry, immunocytochemistry, mass spectrometrometric analyses, e.g ., MALDI-TOF and SELDI-TOF, nucleic acid hybridization techniques, nucleic acid reverse transcription methods, and nucleic acid amplification methods.
  • the level of FR ⁇ not bound to a cell in the sample is assessed by contacting the sample with an antibody that binds FRa.
  • Antibodies that bind FR ⁇ are known in the art and include (i) the murine monoclonal LK26 antibody (the heavy and light chains thereof are presented herein as SEQ ID NOs: 22 and 23), as described in European Patent Application No. 86104170.5 (Rettig) (the entire contents of which are incorporated herein by reference); (ii) the MORAB-003 antibody, as described in International Publication No. WO2004/113388 and U.S. Patent No. 5,646,253 , the entire contents of each of which are incorporated herein by reference.
  • the monoclonal antibodies MOV18 and MOv19 also bind different epitopes on the FR ⁇ molecule (previously known as gp38/FBP). Miotti, S. et al. Int J Cancer, 38: 297-303 (1987 ).
  • the MOV18 antibody binds the epitope set forth herein as SEQ ID NO:26 (TELLNVXMNAK*XKEKPXPX*KLXXQX) (note that at position 12, a tryptophan or histidine residue is possible, and at position 21, an aspartic acid or glutamic acid residue is possible), as taught in Coney et al. Cancer Res, 51: 6125-6132 (1991 ).
  • MORAb-003 refers to an antibody that specifically binds FR ⁇ and which comprises the mature heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the mature light chain sequence of SEQ ID NO:8.
  • the corresponding pre-protein amino acid sequences for MORAb-003 are set forth in SEQ ID NOs: 9 (heavy chain) and 10 (light chain).
  • the MORAb-003 antibody comprises the following CDRs: SEQ ID NO:1 as CDRH1, SEQ ID NO:2 as CDRH2, SEQ ID NO:3 as CDRH3, SEQ ID NO:4 as CDRL1, SEQ ID NO:5 as CDRL2, and SEQ ID NO:6 as CDRL3.
  • MORAb-003 antibody producing cells have been deposited with the American Type Culture Collection (10801 University Boulevard., Manassas, Virginia 20110-2209) on April 24, 2006 and have been assigned Accession No. PTA-7552.
  • antibodies that bind FR ⁇ and for use in the methods of the present invention include 9F3.H9.H3.H3.B5.G2 (also referred to as 9F3),19D4.B7 (also referred to as 19D4), 24F12.B1 (also referred to as 24F12), and 26B3.F2 (also referred to as 26B3).
  • the amino acid sequences of these antibodies, their CDRs, and their heavy and light chain variable domains, as well as polynucleotide sequences that may encode them, are provided in Table 33.
  • these antibodies are murine IgG, or derivatives thereof.
  • the antibodies are human, humanized, or chimeric.
  • the antibody that binds FR ⁇ is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:27.
  • the antibody that binds FR ⁇ includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:28.
  • the antibody that binds FR ⁇ includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO 29.
  • the antibody that binds FR ⁇ includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO 31.
  • the antibody that binds FR ⁇ includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:32. In some embodiments, the antibody that binds FR ⁇ includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:33.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:27; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:28; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:29.
  • the antibody that binds FR ⁇ may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:31; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO: 32; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:33.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:27; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:28; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:29, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:31; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:32; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:33.
  • the antibody that binds FR ⁇ may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:30.
  • the antibody that binds FR ⁇ may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:34.
  • the antibody that binds FR ⁇ may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:30, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:34.
  • the antibody that binds FR ⁇ is the 9F3.H9.H3.H3.B5.G2 (9F3) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FR ⁇ .
  • the antibody has a murine IgG2a constant region.
  • the antibody that binds FR ⁇ is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Boulevard., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11887.
  • the antibody that binds FR ⁇ comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells.
  • antibody that binds FR ⁇ comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • the antibody that binds FR ⁇ is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:35.
  • the antibody that binds FR ⁇ includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:36.
  • the antibody that binds FR ⁇ includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:37.
  • the antibody that binds FR ⁇ includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:39.
  • the antibody that binds FR ⁇ includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:40. In some embodiments, the antibody that binds FR ⁇ includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:41.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:35; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:36; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:37.
  • the antibody that binds FR ⁇ may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:39; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO: 40; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:41.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:35; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:36; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:37, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:39; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:40; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:41.
  • the antibody that binds FR ⁇ may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:38.
  • the antibody that binds FR ⁇ may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:42.
  • the antibody that binds FR ⁇ may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:38, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:42.
  • the antibody that binds FR ⁇ is the 19D4.B7 (19D4) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FR ⁇ .
  • the antibody has a murine IgG2a constant region.
  • the antibody that binds FR ⁇ is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Boulevard., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11884.
  • the antibody that binds FR ⁇ comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells.
  • antibody that binds FR ⁇ comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • the antibody that binds FR ⁇ is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:43.
  • the antibody that binds FR ⁇ includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:44.
  • the antibody that binds FR ⁇ includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:45.
  • the antibody that binds FR ⁇ includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:47.
  • the antibody that binds FR ⁇ includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:48. In some embodiments, the antibody that binds FR ⁇ includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:49.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:43; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:44; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:45.
  • the antibody that binds FR ⁇ may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:47; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO: 48; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:49.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:43; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:44; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:45, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:47; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:48; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:49.
  • the antibody that binds FR ⁇ may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:46.
  • the antibody that binds FR ⁇ may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:50.
  • the antibody that binds FR ⁇ may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:46, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:50.
  • the antibody that binds FR ⁇ is the 24F12.B1 (24F12) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FRa.
  • the antibody has a murine IgG1 constant region.
  • the antibody that binds FR ⁇ is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Boulevard., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11886.
  • the antibody that binds FR ⁇ comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells.
  • antibody that binds FR ⁇ comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • the antibody that binds FR ⁇ is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:51.
  • the antibody that binds FR ⁇ includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:52.
  • the antibody that binds FR ⁇ includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:53.
  • the antibody that binds FR ⁇ includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:55.
  • the antibody that binds FR ⁇ includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:56. In some embodiments, the antibody that binds FR ⁇ includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:57.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:51; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:52; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:53.
  • the antibody that binds FR ⁇ may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:55; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:56; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:57.
  • the antibody that binds FR ⁇ may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:51; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:52; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:53, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:55; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:56; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:57.
  • the antibody that binds FR ⁇ may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:54.
  • the antibody that binds FR ⁇ may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:58.
  • the antibody that binds FR ⁇ may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:54, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:58.
  • the antibody that binds FR ⁇ is the 26B3.F2 (26B3) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FR ⁇ .
  • the antibody has a murine IgG1 constant region.
  • the antibody that binds FR ⁇ is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Boulevard., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11885.
  • the antibody that binds FR ⁇ comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells.
  • antibody that binds FR ⁇ comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • Antigen binding arrangements of CDRs may be engineered using antibody-like proteins as CDR scaffolding. Engineered antigen-binding proteins are included within the scope of antibodies that bind FRa.
  • the antibody that binds FR ⁇ comprises at least one of the following CDRs, as derived from the murine LK26 heavy and light chains: SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. See US Patent No.
  • the antibody includes a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13) LK26HuVKY (SEQ ID NO: 14), LK26HuVKPW (SEQ ID NO: 15), and LK26HuVKPW,Y (SEQ ID NO: 16); and a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). See US Patent No.
  • the antibody comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • the antibody comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • samples may need to be modified in order to make FR ⁇ accessible to antibody binding.
  • slides may be transferred to a pretreatment buffer and optionally heated to increase antigen accessibility. Heating of the sample in the pretreatment buffer rapidly disrupts the lipid bi-layer of the cells and makes the antigens (may be the case in fresh specimens, but not typically what occurs in fixed specimens) ( i.e., the FR ⁇ protein) more accessible for antibody binding.
  • pretreatment buffer are used interchangeably herein to refer to a buffer that is used to prepare cytology or histology samples for immunostaining, particularly by increasing FR ⁇ protein accessibility for antibody binding.
  • the pretreatment buffer may comprise a pH-specific salt solution, a polymer, a detergent, or a nonionic or anionic surfactant such as, for example, an ethyloxylated anionic or nonionic surfactant, an alkanoate or an alkoxylate or even blends of these surfactants or even the use of a bile salt.
  • the pretreatment buffer may, for example, be a solution of 0.1 % to 1% of deoxycholic acid, sodium salt, or a solution of sodium laureth-13-carboxylate ( e.g ., Sandopan LS) or and ethoxylated anionic complex.
  • the pretreatment buffer may also be used as a slide storage buffer.
  • the sample for example, the urine sample, is centrifuged, vortexed, diluted and/or subjected to guanidine treatment.
  • Any method for making FR ⁇ protein more accessible for antibody binding may be used in the practice of the invention, including the antigen retrieval methods known in the art. See, for example, Bibbo, et al. (2002) Acta. Cytol. 46:25-29 ; Saqi, et al. (2003) Diagn. Cytopathol. 27:365-370 ; Bibbo, et al. (2003) Anal. Quant. Cytol. Histol. 25:8-11 , the entire contents of each of which are incorporated herein by reference.
  • samples may be blocked using an appropriate blocking agent, e.g., a peroxidase blocking reagent such as hydrogen peroxide.
  • an appropriate blocking agent e.g., a peroxidase blocking reagent such as hydrogen peroxide.
  • the samples may be blocked using a protein blocking reagent to prevent non-specific binding of the antibody.
  • the protein blocking reagent may comprise, for example, purified casein.
  • An antibody, particularly a monoclonal or polyclonal antibody, that specifically binds to FR ⁇ is then incubated with the sample.
  • Antibody binding to FR ⁇ may be detected through the use of chemical reagents that generate a detectable signal that corresponds to the level of antibody binding and, accordingly, to the level of FR ⁇ protein expression.
  • antibody binding is detected through the use of a secondary antibody that is conjugated to a labeled polymer.
  • labeled polymers include but are not limited to polymer-enzyme conjugates.
  • the enzymes in these complexes are typically used to catalyze the deposition of a chromagen at the antigen-antibody binding site, thereby resulting in cell staining that corresponds to expression level of the biomarker of interest.
  • Enzymes include, but are not limited to, horseradish peroxidase (HRP) and alkaline phosphatase (AP).
  • antibody binding to the FR ⁇ protein is detected through the use of an HRP-labeled polymer that is conjugated to a secondary antibody.
  • Antibody binding can also be detected through the use of a species-specific probe reagent, which binds to monoclonal or polyclonal antibodies, and a polymer conjugated to HRP, which binds to the species specific probe reagent.
  • Slides are stained for antibody binding using any chromagen, e.g., the chromagen 3,3-diaminobenzidine (DAB), and then counterstained with hematoxylin and, optionally, a bluing agent such as ammonium hydroxide or TBS/Tween-20.
  • DAB chromagen 3,3-diaminobenzidine
  • chromagens include, for example, 3-amino-9-ethylcarbazole (AEC).
  • slides are reviewed microscopically by a cytotechnologist and/or a pathologist to assess cell staining, e.g., fluorescent staining (i.e., FR ⁇ expression).
  • cells staining e.g., fluorescent staining (i.e., FR ⁇ expression).
  • samples may be reviewed via automated microscopy or by personnel with the assistance of computer software that facilitates the identification of positive staining cells.
  • the antibody is labeled.
  • detection of antibody binding can be facilitated by coupling the anti-FR ⁇ antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include 125 I, 131 T, 35 S, 14 C, or 3 H.
  • the antibody is labeled with a radio-label, chromophore-label, fluorophore-label, or enzyme-label.
  • frozen samples are prepared as described above and subsequently stained with antibodies against FR ⁇ diluted to an appropriate concentration using, for example, Tris-buffered saline (TBS).
  • Primary antibodies can be detected by incubating the slides in biotinylated anti-immunoglobulin. This signal can optionally be amplified and visualized using diaminobenzidine precipitation of the antigen.
  • slides can be optionally counterstained with, for example, hematoxylin, to visualize the cells.
  • samples are stained with antibodies against FR ⁇ and counterstained as described above for frozen sections.
  • samples may be optionally treated with agents to amplify the signal in order to visualize antibody staining.
  • agents to amplify the signal For example, a peroxidase-catalyzed deposition of biotinyl-tyramide, which in turn is reacted with peroxidase-conjugated streptavidin (Catalyzed Signal Amplification (CSA) System, DAKO, Carpinteria, CA) may be used.
  • CSA Catalyzed Signal Amplification
  • concentration of a particular antibody used to practice the methods of the invention will vary depending on such factors as time for binding, level of specificity of the antibody for FR ⁇ , and method of sample preparation. Moreover, when multiple antibodies are used, the required concentration may be affected by the order in which the antibodies are applied to the sample, e.g., simultaneously as a cocktail or sequentially as individual antibody reagents. Furthermore, the detection chemistry used to visualize antibody binding to FR ⁇ must be optimized to produce the desired signal to noise ratio. In one embodiment of the invention, proteomic methods, e.g., mass spectrometry, are used for detecting and quantitating the FR ⁇ protein.
  • proteomic methods e.g., mass spectrometry
  • MALDI-TOF MS matrix-associated laser desorption/ionization time-of-flight mass spectrometry
  • SELDI-TOF MS surface-enhanced laser desorption/ionization time-of-flight mass spectrometry
  • the present invention is further predicated, at least in part, on the identification of FR ⁇ as a prognostic biomarker, i.e., as a biomarker of the progression and/ or severity, of an FR ⁇ -expressing cancer such as ovarian cancer or non-small cell lung cancer. Accordingly, the present invention provides methods of assessing the progression of an FR ⁇ -expressing cancer in a subject afflicted with ovarian cancer by comparing the level of FR ⁇ in a sample derived from a subject with the level of FR ⁇ in a control sample, wherein a difference in the level of FR ⁇ in the sample (such as a urine or serum sample) derived from the subject compared with the control sample is an indication that the cancer will progress rapidly.
  • a prognostic biomarker i.e., as a biomarker of the progression and/ or severity
  • methods of assessing the level of risk that a subject will develop an FR ⁇ -expressing cancer involve comparing the level of FR ⁇ in a sample derived from a subject with the level of FR ⁇ in a control sample, wherein a difference in the level of FR ⁇ in the sample (such as urine or serum sample) derived from the subject compared with the control sample is an indication that the subject has a higher level of risk of developing an FR ⁇ -expressing cancer as compared to normal risk in a healthy individual.
  • the difference is an increase. In another embodiment, the difference is a decrease. In some types of cancers (e.g ., squamous cell carcinoma of the head and neck, ovarian cancer), a higher level of FR ⁇ expression is associated with a worse prognosis, whereas in other types of cancers (e.g ., non-small-cell lung cancers), a higher level of FR ⁇ expression is associated with a better prognosis.
  • the FR ⁇ -expressing cancer is ovarian cancer or squamous cell carcinoma of the head and neck and the difference is an increase. In another specific embodiment, the FR ⁇ -expressing cancer is a non small-cell lung cancer, and the difference is a decrease.
  • the invention provides methods of assessing the progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer by comparing the level of FR ⁇ in a sample derived from a subject with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the sample (such as a urine or serum sample) derived from the subject compared with the control sample is an indication that the cancer will progress rapidly, or a decrease in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the cancer will progress slowly or will regress.
  • a sample such as a urine or serum sample
  • methods of assessing the level of risk that a subject will develop an FR ⁇ -expressing cancer involve comparing the level of FR ⁇ in a sample derived from a subject with the level of FR ⁇ in a control sample, wherein an increase in the level of FR ⁇ in the sample (such as urine or serum sample) derived from the subject compared with the control sample is an indication that the subject has a higher level of risk of developing an FR ⁇ -expressing cancer as compared to normal risk in a healthy individual, or a decrease in the level of FR ⁇ in the sample derived from the subject as compared with the level of FR ⁇ in the control sample is an indication that the subject has a lower level of risk of developing an FR ⁇ -expressing cancer as compared to a normal risk in a healthy individual.
  • an increase in the level of FR ⁇ in the level of FR ⁇ refers to a level that exceeds a cutoff value determined using an ROC analysis as exemplified in Example 6.
  • a decrease in the level of FR ⁇ refers to a level in a test sample that does not exceed a cutoff value determined using an ROC analysis.
  • the increase or decrease must be greater than the limits of detection of the method for determining the level of FR ⁇ .
  • the increase or decrease be at least greater than the standard error of the assessment method, and preferably a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method.
  • the increase or decrease is assessed using parametric or nonparametric descriptive statistics, comparisons, regression analyses, and the like.
  • the increase or decrease is a level in the sample derived from the subject that is about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900% or about 1000% more or less than the level of FR ⁇ in the control sample.
  • the increase or decrease is a level in the sample derived from the subject that is at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations above or below the average level of FR ⁇ in the control sample.
  • progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer may refer to the progression of an FR ⁇ -expressing cancer from a less severe to a more severe cancer state. This could include an increase in the number or severity of tumors, the degree of metastasis, the speed with which the cancer is growing and spreading, and the like.
  • the progression is a progression from a less severe stage to a more severe stage, wherein the stage is assessed according to a staging scheme known in the art.
  • the FR ⁇ -expressing cancer is ovarian cancer
  • the progression refers to a progression from Stage I to Stage II, from Stage II to Stage III, etc.
  • the progression refers to a progression from Stage 0 to Stage IA, Stage IA to Stage IB, Stage IB to Stage IIA, Stage IIA to Stage IIB, Stage IIB to Stage IIC, etc.
  • the FR ⁇ -expressing cancer is non-small cell lung cancer (NSCLC)
  • the progression refers to a progression from a less severe to a more severe stage as determined under the TNM classification system. See Spira; Greene; Sobin.
  • the phrase "progression of an FR ⁇ -expressing cancer in a subject afflicted with an FR ⁇ -expressing cancer” may refer to a regression of an FR ⁇ -expressing cancer from a more severe state to a less severe state, such as a decrease in the number or severity of tumors, the degree of metastasis, the speed with which the cancer is growing and spreading, and the like.
  • the progression is a progression from a more severe stage to a less severe stage, wherein the stage is assessed according to a staging scheme known in the art.
  • the progression refers to a regression from Stage IV to Stage III, from Stage III to Stage II, etc.
  • the progression refers to a progression from Stage IV to Stage IIIB, Stage IIIB to Stage IIIA, Stage IIIA to Stage IIB, etc.
  • the progression refers to a progression from a more severe to a less severe stage as determined under the TNM classification system. See Spira; Greene; Sobin.
  • the level of FR ⁇ may be used to calculate the likelihood that a subject is afflicted with an FR ⁇ -expressing cancer, the progression of an FR ⁇ -expressing cancer in a subject, the level of risk of developing an FR ⁇ -expressing cancer, the risk of cancer recurrence in a subject being treated for an FR ⁇ -expressing, the survival of a subject being treated for an FR ⁇ -expressing cancer, the efficacy of a treatment regimen for treating an FR ⁇ -expressing cancer, and the like, using the methods of the invention, which may include methods of regression analysis known to one of skill in the art.
  • suitable regression models include, but are not limited to CART ( e.g., Hill, T, and Lewicki, P.
  • a regression analysis includes the level of FR ⁇ .
  • a regression analysis may include additional clinical and/or molecular co-variates.
  • Such clinical co-variates include, but are not limited to, age of the subject, tumor stage, tumor grade, tumor size, treatment regime, e.g ., chemotherapy and/or radiation therapy, clinical outcome (e.g. , relapse, disease-specific survival, therapy failure), and/or clinical outcome as a function of time after diagnosis, time after initiation of therapy, and/or time after completion of treatment.
  • Molecular co-variates can include, but are not limited to additional molecular marker values.
  • markers may include, e.g ., serum CA125 levels, serum DF3 levels, and/or plasma LPA levels.
  • the invention provides methods for monitoring the effectiveness of a therapy or treatment regimen.
  • the present invention provides methods for monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer.
  • the methods involve determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell, in a sample derived from said subject, wherein said subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell with the level of FR ⁇ in a control sample, wherein an increase or no change in the level of FR ⁇ in the sample derived from said subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FR ⁇ in the sample derived from said subject as compared with the level of FR ⁇ in the control sample is an indication that the MORAb-003 treatment is efficacious.
  • FR ⁇ folate receptor alpha
  • control sample may be derived from a subject not subjected to the treatment regimen and a test sample may be derived from a subject subjected to at least a portion of the treatment regimen.
  • test sample and the control sample may be derived from the same subject.
  • the test sample may be a sample derived from a subject after administration of a therapeutic, such as MORAb-003.
  • the control sample may be a sample derived from a subject prior to administration of therapeutic or at an earlier stage of therapeutic regimen. Accordingly, a decrease in the level of expression of FR ⁇ in the test sample, relative to the control sample, is an indication that therapy has decreased the progression of the FR ⁇ -expressing cancer, for example, ovarian cancer.
  • FR ⁇ -expressing cancers wherein a higher level of FR ⁇ is associated with a worse prognosis, such as e.g., ovarian cancer or squamous cell carcinoma of the head and neck
  • a decrease in the level of expression of FR ⁇ in the test sample, relative to the control sample is an indication that therapy is effective in slowing the progression of the FR ⁇ -expressing cancer, or in causing a regression of the cancer, in the subject afflicted with the FR ⁇ -expressing cancer.
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the sample may be urine, serum, plasma or ascites.
  • the sample is urine or serum.
  • the FR ⁇ may be determined by contacting the sample with an antibody that binds FR ⁇ , optionally, using antibodies as described herein and assay methods as described herein.
  • the MORAb-003 treatment antibody is (a) an antibody that comprises the heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the light chain amino acid sequence as set forth in SEQ ID NO:8; (b) an antibody that binds the same epitope as the MORAb-003 antibody; or (c) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • SEQ ID NO:1 GTFSGYGLS
  • SEQ ID NO:2 MISGGSYTYYADSVKG
  • SEQ ID NO:3 HGDDPAWFAY
  • SEQ ID NO:4
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is lung cancer.
  • the lung cancer is non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the NSCLC is selected from the group consisting of adenocarcinoma, squamous cell lung carcinoma, large cell lung carcinoma, pleomorphic NSCLC, carcinoid tumor, salivary gland carcinoma, and unclassified carcinoma.
  • the NSCLC is adenocarcinoma.
  • the lung cancer is small cell lung carcinoma (SCLC).
  • the lung cancer is bronchioalveolar carcinoma.
  • the lung cancer is a lung carcinoid tumor.
  • the invention provides methods of stratifying a subject with an FR ⁇ -expressing cancer into cancer therapy groups based on the determined level of FR ⁇ in a sample.
  • the method involves stratifying a subject with an FR ⁇ -expressing cancer into one of at least four cancer therapy groups.
  • the method involves stratifying a subject with an FR ⁇ -expressing cancer into one of at least about two, about three, about four, about five, about six, about seven, about eight, about nine, or about ten cancer therapy groups.
  • the levels of FR ⁇ may be associated with the severity, i.e., the stage, of the FR ⁇ expressing cancer.
  • ovarian cancer is stratified into different stages based on the severity of the cancer, as set forth herein.
  • the present invention provides methods for stratifying ovarian cancer into Stage I, for example, Stage IA, Stage 1 B or Stage IC; Stage II, for example, Stage IIA, Stage IIB or Stage IIC; Stage III, for example, Stage IIIA, Stage IIIB or Stage IIIC; or Stage IV ovarian cancer.
  • SCLS or NSCLC may be stratified into different stages based on the severity of the cancer, as set forth herein. Accordingly, the present invention provides methods for stratifying the lung cancer, for example, SCLS or NSCLC, into the occult (hidden) stage; stage 0; Stage I, for example, stages IA and IB; Stage II, for example, stages IIA and IIB; Stage III, for example, stages IIIA and IIIB; or Stage IV lung cancer.
  • the present invention is predicated, at least in part, on the finding that FR ⁇ can serve as a predictive biomarker for treatment of FR ⁇ expressing cancers.
  • the methods of the present invention provide for assessing whether a subject will respond to treatment, for example, with MORAb-003, and whether and when to initiate treatment, for example, with MORAb-003, by assessing the levels of FR ⁇ in a subject.
  • the present invention provides a method for predicting whether a subject suffering from an FR ⁇ expressing cancer, for example, ovarian or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in a sample derived from said subject; and comparing the level of folate receptor alpha (FR ⁇ ) which is not bound to a cell in the sample derived from said subject with the level of FR ⁇ in a control sample, wherein a difference between the level of FR ⁇ in the sample derived from said subject and the level of FR ⁇ in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • FR ⁇ folate receptor alpha
  • the degree of difference between the levels of FR ⁇ not bound to a cancer cell in the test sample as compared to the control sample is indicative that the subject will respond to treatment with MORAb-003.
  • a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method is indicative that the subject will respond to treatment with MORAb-003.
  • a difference of at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900% or about 1000% is indicative that the subject will respond to treatment with MORAb-003.
  • a difference of at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations is indicative that the subject will respond to treatment with MORAb-003.
  • the MORAb-003 treatment antibody is (a) an antibody that binds the same epitope as the MORAb-003 antibody; or (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • SEQ ID NO:1 GTFSGYGLS
  • SEQ ID NO:2 MISGGSYTYYADSVKG
  • SEQ ID NO:3 HGDDPAWFAY
  • SEQ ID NO:4 SVSSSISSNNLH
  • SEQ ID NO:5 GTSNLAS
  • SEQ ID NO:6 QQWSSYPYMYT
  • the sample is urine, plasma, serum or ascites.
  • the sample is urine or serum.
  • the FR ⁇ -expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer.
  • the FR ⁇ -expressing cancer is ovarian cancer.
  • the FR ⁇ -expressing cancer is non-small cell lung cancer, such as adenocarcinoma.
  • assay formats known to those of ordinary skill in the art for using an antibody to detect a polypeptide in a sample, including but not limited to enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunofluorimetry, immunoprecipitation, solution phase assay, equilibrium dialysis, immunodiffusion and other techniques. See, e.g ., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988 ; Weir, D. M., Handbook of Experimental Immunology, 1986, Blackwell Scientific, Bost on.
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • the assay may be performed in a Western blot format, wherein a protein preparation from the biological sample is submitted to gel electrophoresis, transferred to a suitable membrane and allowed to react with the antibody. The presence of the antibody on the membrane may then be detected using a suitable detection reagent, as is well known in the art and described below.
  • the assay involves the use of an antibody immobilized on a solid support to bind to the target FR ⁇ polypeptide and remove it from the remainder of the sample.
  • the bound FR ⁇ polypeptide may then be detected using a second antibody reactive with a distinct FR ⁇ polypeptide antigenic determinant, for example, a reagent that contains a detectable reporter moiety.
  • a second antibody reactive with a distinct FR ⁇ polypeptide antigenic determinant for example, a reagent that contains a detectable reporter moiety.
  • the immobilized antibody and the second antibody which recognize distinct antigenic determinants may be any two of the monoclonal antibodies described herein selected from MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein.
  • a competitive assay may be utilized, in which FR ⁇ is labeled with a detectable reporter moiety and allowed to bind to the immobilized anti-FR ⁇ antibody after incubation of the immobilized antibody with the sample.
  • the extent to which components of the sample inhibit the binding of the labeled polypeptide to the antibody is indicative of the reactivity of the sample with the immobilized antibody, and as a result, indicative of the level of FR ⁇ in the sample.
  • the solid support may be any material known to those of ordinary skill in the art to which the antibody may be attached, such as a test well in a microtiter plate, a nitrocellulose filter or another suitable membrane.
  • the support may be a bead or disc, such as glass, fiberglass, latex or a plastic such as polystyrene or polyvinylchloride.
  • the antibody may be immobilized on the solid support using a variety of techniques known to those in the art, which are amply described in the patent and scientific literature.
  • the assay for detection of FR ⁇ in a sample is a two-antibody sandwich assay.
  • This assay may be performed by first contacting a FR ⁇ specific antibody (e.g ., MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein) that has been immobilized on a solid support, commonly the well of a microtiter plate, with the biological sample, such that a soluble molecule naturally occurring in the sample and having an antigenic determinant that is reactive with the antibody is allowed to bind to the immobilized antibody (e.g ., a 30 minute incubation time at room temperature is generally sufficient) to form an antigen-antibody complex or an immune complex.
  • a FR ⁇ specific antibody e.g ., MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein
  • Unbound constituents of the sample are then removed from the immobilized immune complexes.
  • a second antibody specific for FR ⁇ is added, wherein the antigen combining site of the second antibody does not competitively inhibit binding of the antigen combining site of the immobilized first antibody to FR ⁇ (e.g., MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein, that is not the same as the monoclonal antibody immobilized on the solid support).
  • the second antibody may be detectably labeled as provided herein, such that it may be directly detected.
  • the second antibody may be indirectly detected through the use of a detectably labeled secondary (or "second stage") anti-antibody, or by using a specific detection reagent as provided herein.
  • the subject invention method is not limited to any particular detection procedure, as those having familiarity with immunoassays will appreciate that there are numerous reagents and configurations for immunologically detecting a particular antigen (e.g ., FR ⁇ ) in a two-antibody sandwich immunoassay.
  • the first, immobilized antibody specific for FR ⁇ is a polyclonal antibody and the second antibody specific for FR ⁇ is a polyclonal antibody.
  • the first, immobilized antibody specific for FR ⁇ is a monoclonal antibody and the second antibody specific for FR ⁇ is a polyclonal antibody.
  • the first, immobilized antibody specific for FR ⁇ is a polyclonal antibody and the second antibody specific for FR ⁇ is a monoclonal antibody.
  • the first, immobilized antibody specific for FR ⁇ is a monoclonal antibody and the second antibody specific for FR ⁇ is a monoclonal antibody.
  • monoclonal antibodies MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein, as provided herein recognize distinct and noncompetitive antigenic determinants (e.g. , epitopes) on FR ⁇ polypeptides, such that any pairwise combination of these monoclonal antibodies may be employed.
  • the first, immobilized antibody specific for FR ⁇ and/or the second antibody specific for FR ⁇ may be any of the kinds of antibodies known in the art and referred to herein, for example, by way of illustration and not limitation, Fab fragments, F(ab') 2 fragments, immunoglobulin V-region fusion proteins or single chain antibodies.
  • Fab fragments fragments, F(ab') 2 fragments
  • immunoglobulin V-region fusion proteins immunoglobulin V-region fusion proteins or single chain antibodies.
  • the second antibody may contain a detectable reporter moiety or label such as an enzyme, dye, radionuclide, luminescent group, fluorescent group or biotin, or the like.
  • a detectable reporter moiety or label such as an enzyme, dye, radionuclide, luminescent group, fluorescent group or biotin, or the like.
  • the amount of the second antibody that remains bound to the solid support is then determined using a method appropriate for the specific detectable reporter moiety or label. For radioactive groups, scintillation counting or autoradiographic methods are generally appropriate.
  • Antibody-enzyme conjugates may be prepared using a variety of coupling techniques (for review see, e.g., Scouten, W. H., Methods in Enzymology 135:30-65, 1987 ). Spectroscopic methods may be used to detect dyes (including, for example, colorimetric products of enzyme reactions), luminescent groups and fluorescent groups.
  • Biotin may be detected using avidin or streptavidin, coupled to a different reporter group (commonly a radioactive or fluorescent group or an enzyme). Enzyme reporter groups may generally be detected by the addition of substrate (generally for a specific period of time), followed by spectroscopic, spectrophotometric or other analysis of the reaction products. Standards and standard additions may be used to determine the level of mesothelin polypeptide in a sample, using well known techniques.
  • a method of screening for the presence of an FR ⁇ expressing cancer according to the present invention may be further enhanced by the detection of more than one tumor associated marker in a biological sample from a subject. Accordingly, in certain embodiments the present invention provides a method of screening that, in addition to detecting reactivity of FR ⁇ not bound to a cell, also includes detection of at least one additional soluble marker of a malignant condition using established methods as known in the art and provided herein. As noted above, there are currently a number of soluble tumor associated antigens that are detectable in samples of readily obtained biological fluids.
  • kits for assessing whether a subject is afflicted with an FR ⁇ -expressing cancer, for assessing the progression of an FR ⁇ -expressing cancer, for assessing the level of risk that a subject will develop an FR ⁇ -expressing cancer, or for monitoring the effectiveness of a therapy or treatment regimen for an FR ⁇ -expressing cancer include means for determining the level of expression of FR ⁇ and instructions for use of the kit to assess the progression of an FR ⁇ -expressing cancer, to assess the level of risk that a subject will develop an FR ⁇ -expressing cancer, or to monitor the effectiveness of a therapy or treatment regimen for an FR ⁇ -expressing cancer.
  • kits of the invention may optionally comprise additional components useful for performing the methods of the invention.
  • the kits may comprise means for obtaining a sample from a subject, a control sample, e.g., a sample from a subject having slowly progressing cancer and/or a subject not having cancer, one or more sample compartments, and instructional material which describes performance of a method of the invention and tissue specific controls/standards.
  • the means for determining the level of FR ⁇ include known methods in the art for assessing protein levels, as discussed above, and specific preferred embodiments, for example, utilizing the MORAb-003 antibody, as discussed herein.
  • the level of FR ⁇ is assessed by contacting a sample derived from a subject (such as urine or serum) with a folate receptor alpha (FR ⁇ ) binding agent.
  • FR ⁇ folate receptor alpha
  • the binding agent is an antibody.
  • Many of the types of antibodies that bind FR ⁇ are discussed above in the methods of the invention and may also be utilized in the kits of the invention.
  • the means for determining the level of FR ⁇ can further include, for example, buffers or other reagents for use in an assay for determining the level of FR ⁇ .
  • the instructions can be, for example, printed instructions for performing the assay and/or instructions for evaluating the level of expression of FR ⁇ .
  • kits of the inventions may also include means for isolating a sample from a subject.
  • These means can comprise one or more items of equipment or reagents that can be used to obtain a fluid or tissue from a subject.
  • the means for obtaining a sample from a subject may also comprise means for isolating blood components, such as serum, from a blood sample.
  • the kit is designed for use with a human subject.
  • the invention also provides methods (also referred to herein as "screening assays") for identifying modulators, i.e., candidate or test compounds or agents (e.g ., proteins, peptides, peptidomimetics, peptoids, small molecules or other drugs), which modulate the growth, progression and/or aggressiveness of cancer, e.g., an FR ⁇ -expressing cancer, or a cancer cell, e.g., an ovarian cancer cell, by monitoring and comparing the levels of FR ⁇ in a sample.
  • modulators i.e., candidate or test compounds or agents (e.g ., proteins, peptides, peptidomimetics, peptoids, small molecules or other drugs), which modulate the growth, progression and/or aggressiveness of cancer, e.g., an FR ⁇ -expressing cancer, or a cancer cell, e.g., an ovarian cancer cell, by monitoring and comparing the levels of FR ⁇ in a sample.
  • modulators i.e
  • Compounds identified via assays such as those described herein may be useful, for example, for modulating, e.g ., inhibiting, ameliorating, treating, or preventing aggressiveness of an FR ⁇ -expressing cancer or a cancer cell, e.g., an ovarian cancer cell.
  • a cancer cell e.g., an ovarian cancer cell.
  • By monitoring the level of FR ⁇ in a sample one can determine whether the FR ⁇ -expressing cancer is progressing or regressing and whether the test compound has the desired effect. For example, in embodiments wherein the FR ⁇ -expressing cancer is a cancer for which higher levels of FR ⁇ are associated with a worse prognosis, a decrease in the level of FR ⁇ after administration of the test compound(s) would be indicative of the efficacy of the test compound.
  • an increase in the level of FR ⁇ after administration of the test compound(s) would indicate that the test compound is not effective in treating ovarian cancer.
  • the FR ⁇ -expressing cancer is a cancer for which higher levels of FR ⁇ are associated with a better prognosis
  • an increase in the level of FR ⁇ after administration of the test compound(s) would be indicative of the efficacy of the test compound.
  • an decrease in the level of FR ⁇ after administration of the test compound(s) would indicate that the test compound is not effective in treating ovarian cancer.
  • test compounds used in the screening assays of the present invention may be obtained from any available source, including systematic libraries of natural and/or synthetic compounds.
  • Test compounds may also be obtained by any of the numerous approaches in combinatorial library methods known in the art, including biological libraries; peptoid libraries (libraries of molecules having the functionalities of peptides, but with a novel, non-peptide backbone which are resistant to enzymatic degradation but which nevertheless remain bioactive; see, e.g., Zuckermann et al., 1994, J. Med. Chem. 37:2678-85 ); spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound' library method; and synthetic library methods using affinity chromatography selection.
  • the biological library and peptoid library approaches are limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds ( Lam, 1997, Anticancer Drug Des. 12:145
  • EXAMPLE 1 DETERMINATION OF FR ⁇ LEVELS IN URINE SAMPLES FROM HUMAN SUBJECTS WITH AND WITHOUT OVARIAN CANCER AS MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA).
  • Urine samples were obtained from human subjects, including subjects afflicted with ovarian cancer and normal control subjects not afflicted with ovarian cancer.
  • the levels of FR ⁇ in urine samples were determined using an electrochemiluminescence immunoassay (ECLIA) according to the following procedure (see Namba et al. (1999) Analytical Science 15:1087-1093 ):
  • the monoclonal anti-folate receptor alpha antibody was coated over the surface of micro beads (Dynabeads M-450 Epoxy, Dynal). Thirty six milligrams of micro beads were mixed with 1.2 mL of antibody MOV18 (0.36 mg/mL, Enzo Life Science) in 0.15 mol/L phosphate buffer saline pH 7.8 (PBS), followed by gentle mixing for 16 hours at room temperature. The micro beads were then washed 5 times with 50 mM HEPES buffer containing 0.1 % normal rabbit serum (NRS), 150 mmol/L NaCl, 0.01 % Tween 20 pH 7.5 (wash buffer).
  • NBS normal rabbit serum
  • the coated micro beads were suspended in 1.2 mL 50 mM HEPES buffer containing 20% NRS, 150 mmol/L NaCl and 0.01% Tween 20 pH 7.5 (reaction buffer) to block the unbound surface, followed by gentle mixing for 3.5 hours at room temperature. Finally, the micro beads were washed 5 times with wash buffer and re-suspended with 1.2 mL 50 mM HEPES buffer containing 10% NRS, 150 mmol/L NaCl, 10 mmol/L EDTA-2Na and 0.01% Tween 20 pH 7.5 (reaction buffer) so that the concentration of micro beads was 30 mg/mL. The micro beads were stored at 4°C until use.
  • the antibody coated micro beads were set on the reagent table of the Picolumi 8220 (Sanko, Tokyo, Japan) after adjusting the concentration of the beads to 1.5 mg/ml (working solution) in reaction buffer.
  • the Ru labeled antibody was set on the reagent table of the Picolumi 8220 after adjusting the concentration of antibody to 2 ⁇ g/ml (working solution) in reaction buffer.
  • Table 1 depicts the urine levels of FR ⁇ in individual subjects with ovarian cancer and non-afflicted female control subjects.
  • Table 1 FR ⁇ levels in urine of subjects with ovarian cancer and normal female control subjects.
  • Group Sample # FR ⁇ (pg/mL) ovarian cancer 1 27800 2 40242 3 85580 4 4994 5 2017 6 3781 7 29469 8 47456 9 4479 10 11920 11 18352 12 162017 13 30630 14 14431 15 11801 16 13470 17 11563 18 22185 19 52106 normal control 20 8491 21 4885 22 3595 23 21301 24 22757 25 16578 26 6081 27 4195 28 12169 29 20639
  • Figure 2 depicts the distribution of FR ⁇ levels in urine in subjects with ovarian cancer and in normal female control subjects, as set forth in Table 1.
  • Table 2 summarizes the number of subjects (n), mean, standard deviation (SD), maximum (Max.) and minimum (Min.) values for the levels of FR ⁇ in the ovarian cancer group and the normal female control group.
  • Table 2 Summary of urine FR ⁇ measurement FR ⁇ (pg/mL) ovarian cancer normal female N 19 10 Mean 31279 12069 SD 37895 7654 Max. 162017 22757 Min. 2017 3595
  • Dilution Linearity is a measure of accuracy of an assay. Two urine samples were serially diluted by a factor of 10 and 100. The FR ⁇ levels of each sample were measured as set forth in Example 1 and compared to assess the percent error. Percent error was calculated as follows: FR ⁇ in undiluted sample * dilution factor ⁇ FR ⁇ in undiluted sample * 100 FR ⁇ in undiluted sample The results are set forth in Table 3: Table 3: Dilution Linearity for Urine Sample Dilution factor FR ⁇ (pg/mL) Error (%) 1 1 25037 - 10 2601 4 100 279 11 2 1 16649 - 10 1696 2 100 173 4 The foregoing results demonstrate dilution linearity in assessing the levels of FR ⁇ in human urine samples and that, within acceptable errors, urine can be diluted up to a factor of at least 100 while retaining accurate levels of FR ⁇ . Accordingly, dilution of the urine samples may be considered prior to determining levels of FR ⁇ .
  • EXAMPLE 3 CENTRIFUGATION OF URINE SAMPLES - ADDRESSING REPRODUCIBILITY
  • Table 5 depicts the results obtained when three samples were centrifuged prior to performance of the ECLIA assay. Table 5. Reproducibility with sample centrifugation FR ⁇ concentration (ng/mL) Test Sample 1 Sample 2 Sample 3 1 10.4 9.2 13.3 2 10.5 8.9 14.0 3 10.3 9.2 13.4 Mean 10.4 9.1 13.6 SD 0.1 0.1 0.3 CV(%) 1.0 1.1 2.2 As set forth above, the results indicate that centrifugation provided more consistent measurements of FR ⁇ concentration.
  • the levels of FR ⁇ as determined by the ECLIA assay vary depending on whether urine was clarified by centrifugation to remove precipitates or whether urine was vortexed to suspend or disperse sediments. Accordingly, in certain embodiments centrifuging or vortexing of urine samples may be performed prior to determining levels of FR ⁇ .
  • EXAMPLE 4 DETERMINATION OF FR ⁇ LEVELS IN CENTRIFUGED URINE SAMPLES FROM HUMAN SUBJECTS WITH AND WITHOUT OVARIAN CANCER MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA).
  • Example 3 Based on the results of Example 3, the assay for assessing FR ⁇ levels in subjects was modified to introduce a centrifugation step. FR ⁇ levels were determined on the same samples utilized in Example 1, including the group of subjects with ovarian cancer and the group of normal female control subjects.
  • Example 2 The methodology utilized was as described in Example 1, except that the urine samples were centrifuged for 10000 x g for 1 minute and the resulting supernatant subsequently diluted by 1:51 in reaction buffer.
  • Table 7 depicts the levels of FR ⁇ in centrifuged and non-centrifuged urine samples from subjects afflicted with ovarian cancer and healthy female control subjects.
  • Table 7 Urine FR ⁇ 1 level in ovarian cancer and normal control group FR ⁇ (pg/mL) Group Sample # Without centrifugation With centrifugation sediment after centrifugation ovarian cancer 1 27800 23960 + 2 40242 37852 + 3 85580 78976 + 4 4994 3766 + 5 2017 1512 - 6 3781 3443 - 7 29469 25728 + 8 47456 16556 + 9 4479 3357 - 10 11920 5020 + 11 18352 16695 - 12 162017 82705 + 13 30630 4496 + 14 14431 8786 + 15 11801 10582 - 16 13470 5611 + 17 11563 5463 + 18 22185 14443 + 19 52106 38327 - normal control 20 8491 6867 + 21 4885 3754 - 22 3595 3529
  • EXAMPLE 5 DETECTION OF FR ⁇ IN URINE SEDIMENT BY IMMUNOBLOTTING
  • lanes 1-5 correspond to FR ⁇ detected from the following sources:
  • Lane 6 in the western blot represents molecular weight markers and demonstrates that the observed band in lanes 1, 2, 3 and 5 runs at the expected molecular weight for FR ⁇ .
  • Lanes 3 and 5 are positive control samples and lane 4 is a negative control sample.
  • the faint band on lane 1 and the clear band on lane 2 demonstrate that FR ⁇ can be detected in the urine sediment of ovarian cancer patients by western blotting.
  • EXAMPLE 6 DETERMINATION OF FROG LEVELS IN GUANIDINE-TREATED NORMAL HUMAN URINE SAMPLES BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA).
  • Example 4 Based on the results of Example 4 in which centrifugation resulted in decreased FR ⁇ levels, and the results of Example 5 where the urine sediment obtained from centrifugation was shown to contain immunoreactive FR ⁇ , methods were sought to solubilize the sediments of urine to obtain more quantitative and accurate measurements of FR ⁇ .
  • Example 2 The methodology utilized was as described in Example 1, except that the urine samples were mixed in a 1:1 ratio with either 6 M guanidine in buffer (PBS) or buffer alone. Subsequently, the urine samples were diluted by 1:51 in reaction buffer.
  • Table 8 Normal urine FR ⁇ level with or without guanidine treatment Sample Guanidine treatment FR ⁇ (pg/mL) Std Ag Yes 83964 No 82512 Normal Urine 1 Yes 9431 No 7796 2 Yes 5713 No 4066 3 Yes 9687 No 9428 The results of this experiment indicate that guanidine does not interfere with FR ⁇ measurements. As can be seen for the pure antigen control (Std Ag), this methodology of guanidine treatment and subsequent dilution has no effect on the measurement of FR ⁇ . Further, it will be noted that in all three (3) urine samples assessed, the levels of FR ⁇ were higher in the samples treated (solubilized) with guanidine relative to the samples not treated with guanidine.
  • EXAMPLE 7 DETERMINATION OF FR ⁇ LEVELS IN GUANIDINE-TREATED URINE SAMPLES FROM HUMAN SUBJECTS WITH AND WITHOUT OVARIAN CANCER MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA).
  • Example 6 Based on the results of Example 6 in which guanidine treatment was shown not to interfere with FR ⁇ assays, a modified assay protocol was employed to measure FR ⁇ in the urine samples from the subjects with and without ovarian cancer in Example 1.
  • Example 2 The methodology utilized was as described in Example 1, except that the urine samples were mixed in a 1:1 ratio with a 6 M guanidine buffer and subsequently diluted by 1:26 in reaction buffer.
  • Table 10 depicts the levels of FR ⁇ in guanidine treated urine samples from subjects afflicted with ovarian cancer and healthy female control subjects.
  • Table 10 Urine FR ⁇ level in ovarian cancer and normal control group Group Sample # FR ⁇ (pg/mL) ovarian 1 27015 cancer 2 37315 3 79579 4 285 5 1864 6 2902 7 27914 8 51864 9 2699 10 9455 11 18396 12 145564 13 19046 14 10440 15 10977 16 9199 17 18223 18 18747 19 51098 normal control 20 8012 21 3797 22 3323 23 20976 24 6941 25 14512 26 7286 27 2789 28 2617 29 7233
  • Figure 4 shows the distribution of FR ⁇ levels in the urine of ovarian cancer afflicted subjects and normal female control subjects using the modified protocol with guanidine treatment.
  • Table 11 1 summarizes these results.
  • Table 11 Summary of urine FR ⁇ measurement FR ⁇ (pg/mL) ovarian cancer normal control n 19 10 Mean 28557 7749 SD 34990 5850 Max. 145564 20976 Min. 285 2617
  • FIG. 5 shows an ROC curve of the sensitivity and specificity of the ECLIA measurement of FR ⁇ levels in urine after the urine was treated with guanidine.
  • AUC is the area under the curve, which measures the accuracy of the test in separating ovarian cancer from control subjects.
  • the AUC was 0.70 with a positive predictive value of 70% and a negative predictive value of 80%, as shown in Table 12.
  • 15/19 ovarian cancer patients had a concentration of FR ⁇ above 9100 pg/mL and 8/10 normal subjects had a concentration of FR ⁇ less than 9100 pg/mL.
  • Table 12 Guanidine treatment for urine measurement ovarian cancer control Number of Samples 19 10 Positive 15 2 Predictive value (%) 78.9 80.0
  • EXAMPLE 8 CREATININE CORRECTION OF FR ⁇ CONCENTRATIONS DETERMINED IN GUANIDINE-TREATED URINE SAMPLES BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA)
  • FR ⁇ Concentrations of FR ⁇ were previously determined using ECLIA of guanidine-treated urine samples from ovarian cancer patients and normal female controls (See Example 7, Table 10). Here, these FR ⁇ concentrations were corrected for urine creatinine levels in order to normalize for the glomerular filtration rate. The resulting values were subjected to an ROC analysis.
  • Table 13 presents the resulting creatinine-corrected FR ⁇ levels.
  • Table 14 Summary statistics for creatinine-corrected FR ⁇ levels FR ⁇ ( ⁇ g/g-creatinine) ovarian cancer normal control n 18 9 Mean 18.9 6.9 SD 17.9 3.9 Max. 66.0 13.8 Min. 0.6 1.9
  • the creatinine-corrected FR ⁇ levels were further subjected to an ROC analysis.
  • the ROC curve is shown in Figure 7 .
  • Table 15 presents the sensitivity, specificity, and area under the curve (AUC) for various cutoff values of the creatinine-corrected test.
  • EXAMPLE 9 ENZYME IMMUNOASSAY (EIA) AND OPTIMIZATION THEREOF
  • EIA Enzyme Immunoassay
  • the monoclonal anti-folate receptor alpha antibody was coated on the surface of microtiter plates (Nunc-immunoplate, Thermo Scientific) as follows. One hundred microliter of antibody (absorbance 0.02 at 280 nm) in 50 mmol/L carbonate buffer pH 9.4 was dispensed into wells, followed by coating for 16 hours at 4°C. The microplates were then washed 2 times with PBS containing 0.05% Tween20 (PBS-T). Thereafter 0.15 mL of PBS containing 20% normal rabbit serum pH 7.8 was dispensed into wells to block the unbounded surface, followed by blocking for 1 hour at room temperature. Finally, the microplates were washed 2 times with PBS-T. The antibody coated plates were dried and kept at 4°C in aluminum bags until use.
  • Biotin labeling was conducted according to the manufacturers recommendations for the EZ-Link Sulfo-NHS-LC-LC-Biotin (Product No. 21338, Thermo Scientific). Briefly, 1 mg of antibody in 0.4 mL of PBS was mixed with 0.013 mL of 10 mM Sulfo-NHS-LC-LC-Biotin, with an initial molar ratio of antibody to biotin of 1:20, followed by incubation for 30 min at room temperature. The biotin coupled antibody was purified by gel filtration using a PD-10 column (GE Healthcare) eluted with PBS to remove non-reacted biotin. In order to determine the level of biotin incorporation, the EZ Biotin quantitation kit (Product No. 28005, Thermo Scientific) was used. The biotin labeled antibody was stored at -80°C until use.
  • reaction buffer For the first reaction, 40 ⁇ L of plasma or standard antigen and 60 ⁇ L of 50 mM HEPES buffer containing 10% NRS, 150 mmol/L NaCl, 10 mmol/L EDTA-2Na, 0.01% Tween 20 pH 7.5 (reaction buffer) was dispensed into antibody coated wells. The plate was incubated for 18 hours at 4°C, and subsequently washed 5 times with PBS-T. For the second reaction, 100 ⁇ L of 10 ⁇ g/mL biotin labeled antibody in reaction buffer was dispensed. The plate was incubated for 1 hour at room temperature, and subsequently washed 5 times with PBS-T.
  • Figure 8 depicts the EIA assay using MOV18 as the capture antibody and biotinylated MORAb-003 as the detector antibody.
  • biotin labeled antibody and HRP labeled antibody were compared. Compared with HRP labeled antibody, biotin labeled antibody and avidin-HRP provided a higher signal; therefore, biotin labeled antibody and avidin-HRP were employed.
  • the levels of FR ⁇ were measured in human plasma samples taken from ovarian cancer patients and healthy female controls using the electrochemiluminescence assay (ECLIA) described in Example 1 and Figure 1 (using MORAb-003 as the capture antibody and ruthenium (Ru)-labeled MOV-18 as the labeled detector antibody) and the enzyme immunoassay (EIA) described in Example 9 and Figure 8 . In both assays, 40 ⁇ L of plasma was assayed.
  • ELIA electrochemiluminescence assay
  • EIA enzyme immunoassay
  • Table 16 shows the plasma levels of FR ⁇ in ovarian cancer and normal control subjects, as determined using the EIA and ECLIA.
  • Table 16 Plasma concentrations of FR ⁇ determined using EIA and ECLIA methods Group Sample # FR ⁇ concentration (pg/mL) EIA ECLIA Ovarian cancer 1 10 73 2 ⁇ 10 200 3 56 286 4 44 286 5 353 1606 6 83 494 Healthy control 7 110 127 8 162 112 9 88 252 10 180 254 11 262 471 12 206 396
  • EXAMPLE 11 FEASIBILITY OF DIFFERENT TYPES OF ANTIBODIES FOR EIA
  • the levels of FR ⁇ in plasma from ovarian cancer patients and normal healthy female controls were measured using an enzyme-linked immunosorbent assay (EIA) with different combinations of capture and biotin-labeled antibodies and compared with the levels of FR ⁇ measured using the ECLIA assay.
  • EIA enzyme-linked immunosorbent assay
  • the ECLIA method was as described in Example 1 and depicted in Figure 1 (using the MORAb-003 antibody as the capture antibody and the Mov-18 antibody as the labeled detector antibody).
  • the EIA method was as described in Example 9, except that three different combinations of capture/detector antibodies were employed, as depicted in Figure 10 : MOV18/MORAb-003, 548908/MORAb-003 and 6D398/MORAb-003.
  • the antibodies 548908 and 6D398 are commercially available.
  • the 548908 antibody was obtained from R&D Systems (North Las Vegas, NV) and the 6D398 antibody was obtained from US Biological (Swampscott, MA 01907).
  • the concentrations of FR ⁇ (pg/mL) determined using the EIA and ECLIA methods are shown in Table 18.
  • concentrations of FR ⁇ (pg/ml) determined by EIA using various combinations of capture/detector antibodies are depicted graphically in Figure 11 .
  • Table 18 Plasma concentrations of FR ⁇ (pg/mL) determined using the EIA and ECLIA methods with various combinations of capture and detector antibodies.
  • EXAMPLE 12 PLASMA LEVELS OF FR ⁇ DETERMINED By EIA AND ECLIA IN SAMPLES FROM OVARIAN CANCER PATIENTS
  • Table 20 Plasma FR ⁇ concentrations in ovarian cancer patients and normal controls, as determined using EIA and ECLIA Group Sample # EIA ECLIA pg/mL pg/mL Ovarian cancer 1 245 217 2 247 223 3 194 229 4 2613 1335 5 154 153 6 319 215 7 516 390 8 370 271 9 933 449 10 4768 4502 11 385 266 12 251 322 13 404 349 14 338 371 15 4147 2344 16 179 165 17 380 296 Control 18 232 181 19 372 173 20 332 189 21 380 203 22 376 290 23 406 217 24 281 182 25 348 191 26 490 247 27 253 137 28 368 185 29 338 195 30 289 219 31 338 206 32 406 226 33 365 228 34 501 280 35 806 388 36 613 286 37 380 250 38 420 281 39 393 280 40 552 2
  • Figure 13 shows the distribution of plasma FR ⁇ concentrations in subjects with ovarian cancer and normal female control subjects as determined using EIA.
  • Table 21 shows summary descriptive statistics for the plasma FR ⁇ concentrations in ovarian cancer and normal female control subjects as determined using EIA. Table 21: Summary of FR ⁇ plasma concentrations in ovarian cancer and normal female control subjects as determined using EIA.
  • EXAMPLE 13 DETERMINATION OF FR ⁇ LEVELS IN MATCHED URINE AND SERUM SAMPLES FROM LUNG CANCER PATIENTS AND OVARIAN CANCER PATIENTS AS MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA)
  • FR ⁇ levels were determined in matched urine and serum samples from lung cancer and ovarian cancer patients using ECLIA where the samples were taken from the same patient. The correlation between serum and urine FR ⁇ levels was also determined.
  • Table 22 The results of the ECLIA assays of serum and urine from lung cancer and ovarian cancer patients are presented in Table 22.
  • Table 22 FR ⁇ concentrations in matched urine and serum samples of lung cancer patients and ovarian cancer patients, as determined by ECLIA Serum Urine Group set ID pg/mL pg/mL Lung cancer 1 146 2009 2 153 4496 3 206 - 4 70 3562 5 195 12381 6 352 21873 7 198 11296 8 120 18570 9 275 4455 10 163 8662 11 145 5294 12 178 - 13 165 1106 14 187 7446 15 168 11167 16 217 24448 17 142 6724 18 177 14514 19 236 822 20 101 4826 21 145 7723 22 213 9887 23 143 7422 24 253 3376 25 421 8045 ovarian cancer 26 282 9414 27 1605 7651 28 240 13059 29 695 10549
  • Table 23 Summary statistics for FR ⁇ concentrations in matched serum and urine samples of lung cancer patients, as determined by ECLIA Lung cancer FR ⁇ (pg/mL) Serum Urine n 25 23 Mean 191 8700 SD 75 6291 Max. 421 24448 Min. 70 822
  • Table 24 Summary statistics for FR ⁇ concentrations in matched serum and urine samples of ovarian cancer patients, as determined by ECLIA Ovarian cancer FR ⁇ (pg/mL) Serum Urine n 4 4 Mean 705 10168 SD 634 2266 Max. 1605 13059 Min. 240 7651
  • Figure 15 shows correlations between ECLIA measures of FR ⁇ levels in matched serum and urine samples taken from the same patient.
  • EXAMPLE 14 ASSESSMENT OF LEVELS OF FR ⁇ IN SERUM SAMPLES FROM PATIENTS WITH OVARIAN CANCER, PATIENTS WITH LUNG CANCER, AND NORMAL CONTROLS
  • Serum FR ⁇ levels in the serum of patients with ovarian cancer, patients with lung cancer, and normal controls were assessed. Serum FR ⁇ levels were assessed using ECLIA with two different pairs of capture-detector antibodies: Pair 1, in which 9F3 was the capture antibody and 24F12 was the detector antibody, and Pair 2, in which 26B3 was the capture antibody and 19D4 was the detector antibody.
  • the FR ⁇ pairs were tested with full calibrator curves and 196 individual serums diluted 1:4.
  • 26B3 was used as the capture antibody after CR processing on a plate lot (75 ⁇ g/mL, +B, +T) and 19D4 was used as the detector antibody at 1.0 ⁇ g/mL.
  • 9F3 was used as the capture antibody and 24F12 was used as the detector antibody at 1.0 ⁇ g/mL.
  • Diluent 100 Meso Scale Discovery, Gaithersburg, Maryland
  • HAMA human anti-mouse antibody
  • Diluent 3 (Meso Scale Discovery, Gaithersburg, Maryland) was used for detections.
  • Table 25 FR ⁇ levels in serum of patients with ovarian cancer, patients with lung cancer, and normal controls
  • LLOQ 1 1pg/m
  • LLOQ 1 5 pg/mL FR ⁇ - Pair 2 FR ⁇ Pair 1
  • MSD Sample Testing Number Sample Type Adjusted backfit conc 2 (pg/mL) %CV Adjusted backfit conc 2 (pg/mL) %CV stage grade gender comments 1 Ovarian Serum 3760 4% 3585 4% III 2 F Adenocarcinoma-Ovary 2 Ovarian Serum 223 3% 273 2% III 3 F Aderiocarcinoma-Ovary 3 Ovarian Serum 950 1% 3346 8% IIIC F Papillary Serous Carcinoma 4 Ovarian Serum 3827 4% 968 0% III 2 F Adenocarcinoma-Ovary 5 Ovarian Serum 251 6% 468 2% IV 2 F Aderiocarcinoma-Ovary
  • 9F3, 2412, 26B3 and 19D4 antibodies were useful in detecting levels of FR ⁇ in biological samples, for example serum, derived from a subject.
  • biological samples for example serum, derived from a subject.
  • the particular combinations of (i) 9F3 as a capture antibody and 24F12 as a detector antibody and (ii) 26B3 as a capture antibody and 19D4 as a detector antibody were capable and particularly effective of assessing levels of FR ⁇ in biological samples.
  • the ability of three anti-FR ⁇ antibody pairs in detecting the levels of FR ⁇ in urine samples was assessed.
  • the antibody pairs utilized were as follows: (1) 26B3 as detector antibody and 9F3 as capture antibody, and (2) 24F12 as detector antibody and 9F3 as capture antibody.
  • Table 28 Detection of FR ⁇ levels in urine using 26B3 as detector antibody and 9F3 as capture antibody Detect 26B3 Capture 9F3 3M Guanidine PBS Control Sample ID Adjusted Backfit conc pg/mL %CV % of control Adjusted Backfit conc pg/mL %CV Urine Ind 2 33,824 4% 98% 34,569 2% Urine Ind 3 2,086 4% 99% 2,107 3% Urine Ind 4 15,283 5% 97% 15,696 2% Urine Ind 5 24,955 4% 92% 26,991 3% Average 4% 97% Average 3% Table 29: Detection of FR ⁇ levels in urine using 24F12 as detector antibody and 9F3 as capture antibody Detect 24F12 Capture 9F3 3M Guanidine PBS Control Sample ID Adjusted Backfit conc pg/mL %CV % of control Adjusted Backfit conc pg/mL %CV Urine Ind 2 38,455 4%
  • results of this second set of experiments further confirm the results of the first set of experiments and demonstrate that the level of FR ⁇ which is not bound to a cell can be reliably assessed, for example, in urine, using assays such as the ECLIA assay and using the 26B3, 9F3, 24F12 antibodies. Further, the results demonstrate that such assays can effectively detect FR ⁇ using pairs of detector and capture antibodies that bind FR ⁇ (such as, e.g., 26B3 as detector antibody and 9F3 as capture antibody, and 24F12 as detector antibody and 9F3 as capture antibody).
  • the levels of FR ⁇ were assessed in samples of serum and plasma on two separate days.
  • the subjects from whom the samples were derived were either normal subjects or patients with ovarian or lung cancer.
  • the protocol for assessing FR ⁇ levels was the same as set forth in Example 14 above.
  • the pairs of antibodies used for assessing FR ⁇ levels were also the same as in Example 14, i.e., Pair 1, in which 9F3 was the capture antibody and 24F12 was the detector antibody, and Pair 2, in which 26B3 was the capture antibody and 19D4 was the detector antibody.
  • Table 30 Levels of FR ⁇ as assessed in serum and plasma samples on different days Day 1 Day 2 Day 1 Day 2 Day 1 Day 2 Day 1 Day 2 FR ⁇ /pair1 FR ⁇ /pair1 FR ⁇ /pair1 FR ⁇ /pair1 FR ⁇ /pair2 FR ⁇ /pair2 FR ⁇ /pair2 FR ⁇ /pair2 Donor ID Disease Biosample Confirmed Diagnosis Stage Serum (pg/ml) Serum (pg/ml) Plasma (pg/ml) Plasma (pg/ml) Serum (pg/ml) Serum (pg/ml) Plasma (pg/mL) Plasma (pg/mL) 17168 ovary Serum FRA - Pair 1 versus Pair 2 I 1236 1282 1466 1183 1298 1384 1410 1336 46464 ovary Serous carcinoma IIIC 1589 1848 2027 2147 1966 2018 2066 2210 47219 ovary Adenocarcinom
  • 9F3, 2412, 26B3 and 19D4 antibodies were useful in detecting levels of FR ⁇ in biological samples, for example, serum or plasma, derived from a subject.
  • biological samples for example, serum or plasma
  • the particular combinations of (i) 9F3 as a capture antibody and 24F12 as a detector antibody and (ii) 26B3 as a capture antibody and 19D4 as a detector antibody were capable and particularly effective in assessing levels of FR ⁇ in biological samples.
  • Figure 16 shows the correlation in serum versus plasma FR ⁇ levels for assays conducted using Pair 1 (see Example 16). The R 2 value was 0.8604.
  • Figure 17 shows the correlation in serum versus plasma FR ⁇ levels for assays conducted using Pair 2 (see Example 16). The R 2 value was 0.9766.
  • Figure 18 shows the correlation in serum FR ⁇ levels for assays conducted using Pair 1 versus Pair 2 (see Example 16). The R 2 value was 0.9028.
  • Figure 19 shows the correlation in plasma FR ⁇ levels for assays conducted using pair 1 versus pair 2 (see Example 16). The R 2 value was 0.8773.
  • Figure 20 shows the interday correlation in serum FR ⁇ levels for assays conducted using pair 2.
  • the R 2 value was 0.9839.
  • Table 33 SEQUENCES SEQ ID NO: DESCRIPTION SEQUENCE 1 MORAB-003 CDRH1 GFTFSGYGLS 2 MORAB-003 CDRH2 MISSGGSYTYYADSVKG 3 MORAB-003 CDRH3 HGDDPAWFAY 4 MORAB-003 CDRL1 SVSSSISSNNLH 5 MORAB-003 CDRL2 GTSNLAS 6 MORAB-003 CDRL3 QQWSSYPYMYT 7 MORAb-003 Heavy Chain Mature Polypeptide Amino Acid Sequence 8 MORAb-003 Light Chain Mature Polypeptide Amino Acid Sequence 9 MORAb-003 Heavy Chain full length pre-protein amino acid sequence 10 MORAb-003 Light Chain full length pre-protein amino acid sequence 11 MORAb-003 Heavy Chain Nucleotide 12 MORAb-003 Light Chain Nucleotide 13 LK26HuVK 14 LK26HuVKY 15 LK26HuVKPW 16 LK26

Abstract

The present invention provides methods and kits for assessing whether a subject is afflicted with an FRα-expressing cancer, methods and kits for predicting the progression of ovarian cancer in a subject afflicted with an FRα-expressing cancer, methods and kits for assessing the level of risk that a subject will develop an FRα-expressing cancer, and methods of stratifying a subject with an FRα-expressing cancer into cancer therapy groups. The methods involve determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject and comparing this level with the level of FRα in a control sample.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of the filing date of U.S. Provisional Application No. 61/410,497, filed on November 5, 2010 , and U.S. Provisional Application No. 61/508,444, filed on July 15, 2011 , the entire contents of each of which are hereby incorporated by reference.
  • BACKGROUND OF THE INVENTION
  • In humans, the high affinity receptor for folate comes in three isoforms: alpha, beta, and gamma. The alpha and beta forms are typically bound to the membranes of cells by a glycosyl phosphatidylinositol (GPI) anchor. They recycle between extracellular and endocytic compartments and are capable of transporting folate into the cell. Soluble forms of FRα may be derived by the action of proteases or phospholipase on membrane anchored folate receptors.
  • Folate receptor alpha (also referred to as FRα, FR-alpha, FOLR-1 or FOLR1) is expressed in a variety of epithelial tissues, including those of the choroid plexus, lung, thyroid, kidney, uterus, breast, Fallopian tube, epididymis, and salivary glands. Weitman, SD et al. Cancer Res 52: 3396-3401 (1992); Weitman SD et al, Cancer Res 52: 6708-6711. Overexpression of FRα has been observed in various cancers, including lung cancer (e.g., bronchioalveolar carcinomas, carcinoid tumors, and non-small cell lung cancers, such as adenocarcinomas); mesothelioma; ovarian cancer; renal cancer; brain cancer (e.g., anaplastic ependymoma, cerebellar juvenile pilocytic astrocytoma, and brain metastases); cervical cancer; nasopharyngeal cancer; mesodermally derived tumor; squamous cell carcinoma of the head and neck; endometrial cancer; endometrioid adenocarcinomas of the ovary, serous cystadenocarcinomas, breast cancer; bladder cancer; pancreatic cancer; bone cancer (e.g., high-grade osteosarcoma); pituitary cancer (e.g., pituitary adenomas); colorectal cancer and medullary thyroid cancer. See e.g., U.S. Patent No. 7,754,698 ; U.S. Patent Application No. 2005/0232919 ; WO 2009/132081 ; Bueno R et al. J of Thoracic and Cardiovascular Surgery, 121(2) : 225-233 (2001); Elkanat H & Ratnam M. Frontiers in Bioscience, 11, 506-519 (2006); Fisher R.E. J Nucl Med, 49: 899-906 (2008); Franklin, WA et al. Int J Cancer, Suppl 8: 89-95 (1994); Hartman L.C. et al. Int J Cancer 121: 938-942 (2007); Iwakiri S et al. Annals of Surgical Oncology, 15(3): 889-899; Parker N. et al. Analytical Biochemistry, 338: 284-293 (2005); Weitman, SD et al. Cancer Res 52: 3396-3401 (1992); Saba N.F. et al. Head Neck, 31(4): 475-481 (2009); Yang R et al. Clin Cancer Res 13: 2557-2567 (2007). In some types of cancers (e.g., squamous cell carcinoma of the head and neck), a higher level of FRα expression is associated with a worse prognosis, whereas in other types of cancers (e.g., non-small-cell lung cancers), a higher level of FRα expression is associated with a better prognosis. See, e.g., Iwakiri S et al. Annals of Surgical Oncology, 15(3): 889-899; Saba N.F. et al. Head Neck, 31(4): 475-481 (2009).
  • Earlier detection of cancer improves survival rates and quality of life. To improve the likelihood of early detection and treatment, a pressing need exists for non-invasive methods for diagnosing cancer, for determining the level of risk of developing cancer, and for predicting the progression of cancer. The present invention satisfies these needs for FRα-expressing cancers.
  • SUMMARY OF THE INVENTION
  • The present invention provides methods of assessing whether a subject is afflicted with FRα-expressing cancers such as lung or ovarian cancer, methods of assessing the progression of FRα-expressing cancers such as lung or ovarian cancer in a subject afflicted with the FRα-expressing cancers, methods of stratifying an FRα-expressing cancer subject into one of at least four cancer therapy groups, methods of assessing the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer and kits for assessing whether a subject is afflicted with FRα-expressing cancers such as lung or ovarian cancer or for assessing the progression of FRα-expressing cancers such as lung or ovarian cancer in a subject.
  • Methods of Assessing Whether a Subject is Afflicted with an FRα Expressing Cancer
  • In a first aspect, the present invention provides a method of assessing whether a subject is afflicted with an FRα-expressing cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein a difference between the level of FRα in the sample derived from the subject and the level of FRα in the control sample is an indication that the subject is afflicted with an FRα-expressing cancer; wherein the level of FRα in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FRα. In a particular embodiment, the sample is either urine, serum, plasma or ascites.
  • In another aspect, the present invention is directed to a method of assessing whether a subject is afflicted with an FRα-expressing cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the urine sample derived from the subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the urine sample derived from the subject and the level of FRα in the control sample is an indication that the subject is afflicted with an FRα-expressing cancer. In a further aspect, the present invention provides a method of assessing whether a subject is afflicted with a cancer that expresses FRα, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the serum sample derived from the subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the serum sample derived from the subject and the level of FRα in the control sample is an indication that the subject is afflicted with an FRα-expressing cancer.
  • In various embodiments of the foregoing aspects of the invention, the FRα-expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer. In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In another embodiment, the FRα-expressing cancer is non-small cell lung cancer, such as an adenocarcinoma.
  • In another aspect, the present invention is directed to methods of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FRα which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer.
  • In various aspects of the foregoing aspects of the invention, the presence of FRα in the urine sample at a concentration of greater than about 9500 pg/mL, about 10,000 pg/mL, about 11,000 pg/mL, about 12,000 pg/mL, about 13,000 pg/mL, about 14,000 pg/mL, about 15,000 pg/mL, about 16,000 pg/mL, about 17,000 pg/mL, about 18,000 pg/mL, about 19,000 pg/mL, or about 20,000 pg/mL is an indication that the subject is afflicted with ovarian cancer.
  • In various aspects, the level of FRα is determined by contacting the sample with an antibody that binds FRα. For example, the antibody is selected from the group consisting of:
    1. (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    2. (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    3. (c) the MOV18 antibody;
    4. (d) an antibody that binds the same epitope as the MOV18 antibody;
    5. (e) the 548908 antibody;
    6. (f) an antibody that binds the same epitope as the 548908 antibody;
    7. (g) the 6D398 antibody;
    8. (h) an antibody that binds the same epitope as the 6D398 antibody;
    9. (i) an antibody that binds the same epitope as the 26B3 antibody;
    10. (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    11. (k) the 26B3 antibody;
    12. (l) an antibody that binds the same epitope as the 19D4 antibody;
    13. (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    14. (n) the 19D4 antibody;
    15. (o) an antibody that binds the same epitope as the 9F3 antibody;
    16. (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    17. (q) the 9F3 antibody;
    18. (r) an antibody that binds the same epitope as the 24F12 antibody;
    19. (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    20. (t) the 24F12 antibody;
    21. (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    22. (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    23. (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    24. (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    25. (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the antibody binds the same epitope as the MORAb-003 antibody. In another embodiment, the antibody includes SEQ ID NO: 1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. In another embodiment, the antibody is the MOV18 antibody. In yet another embodiment, the antibody binds the same epitope as the MOV18 antibody. In a further embodiment, the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16). Alternatively or in combination, the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). In certain embodiments, the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the level of FRα in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV 18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • In certain embodiments, the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody. Alternatively, or in combination, the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • In certain embodiments, the level of FRα is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • In various embodiments of the foregoing aspects of the invention, the control sample is a standardized control level of FRα in a healthy subject.
  • In certain embodiments, the sample is treated with guanidine prior to determining the level of FRα in the sample. Alternatively or in combination, the sample is diluted prior to determining the level of FRα in the sample. Alternatively, or in combination, the sample is centrifuged, vortexed, or both, prior to determining the level of FRα in the sample.
  • In yet another aspect, the present invention is directed to a method of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the sample with the level of FRα in a control sample, wherein a difference between the levels of FRα in the sample derived from the subject and in the control sample is an indication that the subject is afflicted with ovarian cancer; wherein the level of FRα in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody. For example, the sample may be urine, serum, plasma or ascites.
  • Methods of Assessing the Progression of an FRα Expressing Cancer in a subject
  • In a further aspect, the present invention is directed to a method of assessing the progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein an increase in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the cancer will progress rapidly; and wherein a decrease in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the cancer will progress slowly or will regress, thereby assessing the progression of the FRα-expressing cancer in the subject; wherein the level of FRα which is not bound to a cell in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FRα. In a particular embodiment, the sample is urine, serum, plasma or ascites.
  • In another aspect, the present invention provides a method of assessing the progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the urine sample derived from the subject with the level of FRα in a control sample, wherein an increase in the level of FRα in the urine sample derived from the subject as compared with the level of FRα in the control sample is an indication that the cancer will progress rapidly; and wherein a decrease in the level of FRα in the urine sample derived from the subject as compared with the level of FRα in the control sample is an indication that the cancer will progress slowly or will regress, thereby assessing the progression of the FRα-expressing cancer in the subject.
  • In a further aspect, the present invention provides methods of assessing the progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the serum sample derived from the subject with the level of FRα in control sample, wherein an increase in the level of FRα in the serum sample derived from the subject as compared with the level of FRα in the control sample is an indication that the cancer will progress rapidly; and wherein a decrease in the level of FRα in the serum sample derived from the subject as compared with the level of FRα in the control sample is an indication that the cancer will progress slowly or will regress, thereby assessing the progression of the FRα-expressing cancer in the subject.
  • In various embodiments of the foregoing aspects of the invention, the FRα-expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer. In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In another embodiment, the FRα-expressing cancer is non-small cell lung cancer, such as an adenocarcinoma.
  • In another aspect, the present invention is directed to methods of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FRα which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer.
  • In various aspects of the foregoing aspects of the invention, the presence of FRα in the urine sample at a concentration of greater than about 9500 pg/mL, about 10,000 pg/mL, about 11,000 pg/mL, about 12,000 pg/mL, about 13,000 pg/mL, about 14,000 pg/mL, about 15,000 pg/mL, about 16,000 pg/mL, about 17,000 pg/mL, about 18,000 pg/mL, about 19,000 pg/mL, or about 20,000 pg/mL is an indication that the subject is afflicted with ovarian cancer.
  • In various aspects, the level of FRα is determined by contacting the sample with an antibody that binds FRα. For example, the antibody is selected from the group consisting of:
    1. (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    2. (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    3. (c) the MOV18 antibody;
    4. (d) an antibody that binds the same epitope as the MOV18 antibody;
    5. (e) the 548908 antibody;
    6. (f) an antibody that binds the same epitope as the 548908 antibody;
    7. (g) the 6D398 antibody;
    8. (h) an antibody that binds the same epitope as the 6D398 antibody;
    9. (i) an antibody that binds the same epitope as the 26B3 antibody;
    10. (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    11. (k) the 26B3 antibody;
    12. (l) an antibody that binds the same epitope as the 19D4 antibody;
    13. (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    14. (n) the 19D4 antibody;
    15. (o) an antibody that binds the same epitope as the 9F3 antibody;
    16. (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    17. (q) the 9F3 antibody;
    18. (r) an antibody that binds the same epitope as the 24F12 antibody;
    19. (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    20. (t) the 24F12 antibody;
    21. (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    22. (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    23. (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    24. (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    25. (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the antibody binds the same epitope as the MORAb-003 antibody. In another embodiment, the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. In another embodiment, the antibody is the MOV 18 antibody. In yet another embodiment, the antibody binds the same epitope as the MOV18 antibody. In a further embodiment, the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16). Alternatively or in combination, the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). In certain embodiments, the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the level of FRα in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • In certain embodiments, the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody. Alternatively, or in combination, the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • In certain embodiments, the level of FRα is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • In various embodiments of the foregoing aspects of the invention, the control sample is a standardized control level of FRα in a healthy subject. In another embodiment, the control sample is a sample previously obtained from the subject.
  • In certain embodiments, the sample is treated with guanidine prior to determining the level of FRα in the sample. Alternatively or in combination, the sample is diluted prior to determining the level of FRα in the sample. Alternatively, or in combination, the sample is centrifuged, vortexed, or both, prior to determining the level of FRα in the sample.
  • In a further aspect, the present invention provides methods of assessing the progression of ovarian cancer in a subject afflicted with ovarian cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the sample with the level of FRα in a control sample, wherein an increase in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the ovarian cancer will progress rapidly; and wherein a decrease in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the ovarian cancer will progress slowly or will regress, thereby assessing the progression of ovarian cancer in the subject; wherein the level of FRα in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody. For example, the sample may be urine, serum, plasma or ascites.
  • Methods of Stratifying an FRα Epressing Cancer into Cancer Therapy Groups
  • In a further aspect, the present invention provides a method of stratifying a subject afflicted with an FRα-expressing cancer into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FRα) which is not bound to a cell; wherein the level of FRα which is not bound to a cell in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FRα. For example, the sample is selected from the group consisting of urine, serum, plasma or ascites.
  • In yet another aspect, the present invention provides a method of stratifying a subject afflicted with an FRα-expressing cancer into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FRα) which is not bound to a cell in the sample. In a further aspect, the present invention is directed to methods of stratifying a subject afflicted with an FRα-expressing cancer into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a serum sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FRα) which is not bound to a cell in the serum sample.
  • In various embodiments of the foregoing aspects of the invention, the FRα-expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer. In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In another embodiment, the FRα-expressing cancer is non-small cell lung cancer, such as an adenocarcinoma.
  • In another aspect, the present invention is directed to methods of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FRα which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer.
  • In various aspects of the foregoing aspects of the invention, the presence of FRα in the urine sample at a concentration of greater than about 9500 pg/mL, about 10,000 pg/mL, about 11,000 pg/mL, about 12,000 pg/mL, about 13,000 pg/mL, about 14,000 pg/mL, about 15,000 pg/mL, about 16,000 pg/mL, about 17,000 pg/mL, about 18,000 pg/mL, about 19,000 pg/mL, or about 20,000 pg/mL is an indication that the subject is afflicted with ovarian cancer.
  • In various aspects, the level of FRα is determined by contacting the sample with an antibody that binds FRα. For example, the antibody is selected from the group consisting of:
    1. (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    2. (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    3. (c) the MOV18 antibody;
    4. (d) an antibody that binds the same epitope as the MOV18 antibody;
    5. (e) the 548908 antibody;
    6. (f) an antibody that binds the same epitope as the 548908 antibody;
    7. (g) the 6D398 antibody;
    8. (h) an antibody that binds the same epitope as the 6D398 antibody;
    9. (i) an antibody that binds the same epitope as the 26B3 antibody;
    10. (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    11. (k) the 26B3 antibody;
    12. (l) an antibody that binds the same epitope as the 19D4 antibody;
    13. (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    14. (n) the 19D4 antibody;
    15. (o) an antibody that binds the same epitope as the 9F3 antibody;
    16. (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    17. (q) the 9F3 antibody;
    18. (r) an antibody that binds the same epitope as the 24F12 antibody;
    19. (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    20. (t) the 24F12 antibody;
    21. (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    22. (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    23. (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    24. (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    25. (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the antibody binds the same epitope as the MORAb-003 antibody. In another embodiment, the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. In another embodiment, the antibody is the MOV18 antibody. In yet another embodiment, the antibody binds the same epitope as the MOV 18 antibody. In a further embodiment, the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16). Alternatively or in combination, the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). In certain embodiments, the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the level of FRα in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • In certain embodiments, the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody. Alternatively, or in combination, the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • In certain embodiments, the level of FRα is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • In various embodiments of the foregoing aspects of the invention, the control sample is a standardized control level of FRα in a healthy subject.
  • In certain embodiments, the sample is treated with guanidine prior to determining the level of FRα in the sample. Alternatively or in combination, the sample is diluted prior to determining the level of FRα in the sample. Alternatively, or in combination, the sample is centrifuged, vortexed, or both, prior to determining the level of FRα in the sample.
  • In a particular embodiment, the subject is stratified in Stage I, Stage II, Stage III or Stage IV ovarian cancer.
  • In a further aspect, the present invention provides a method of stratifying an ovarian cancer subject into one of at least four cancer therapy groups by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject; and stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FRα) which is not bound to a cell in the sample; wherein the level of FRα in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody. For example, the sample may be urine, serum, plasma or ascites.
  • Methods of Monitoring the Efficacy of MORAb-003 Treatment of Ovarian Cancer or Lung Cancer
  • In one aspect, the present invention provides a method of monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from the subject, wherein the subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein an increase in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is efficacious. In particular embodiments, the level of FRα which is not bound to a cell in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FRα. For example, the sample may be urine, serum, plasma or ascites.
  • In a further aspect, the present invention provides a method of monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject, wherein the subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the urine sample derived from the subject with the level of FRα in a control sample, wherein an increase in the level of FRα in the urine sample derived from the subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FRα in the urine sample derived from the subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is efficacious.
  • In yet another aspect, the present invention is directed to a method of monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a serum sample derived from the subject, wherein the subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the serum sample derived from the subject with the level of FRα in a control sample, wherein an increase in the level of FRα in the serum sample derived from the subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FRα in the serum sample derived from the subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is efficacious.
  • In various aspects, the level of FRα is determined by contacting the sample with an antibody that binds FRα. For example, the antibody is selected from the group consisting of:
    1. (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    2. (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    3. (c) the MOV18 antibody;
    4. (d) an antibody that binds the same epitope as the MOV18 antibody;
    5. (e) the 548908 antibody;
    6. (f) an antibody that binds the same epitope as the 548908 antibody;
    7. (g) the 6D398 antibody;
    8. (h) an antibody that binds the same epitope as the 6D398 antibody;
    9. (i) an antibody that binds the same epitope as the 26B3 antibody;
    10. (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    11. (k) the 26B3 antibody;
    12. (l) an antibody that binds the same epitope as the 19D4 antibody;
    13. (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    14. (n) the 19D4 antibody;
    15. (o) an antibody that binds the same epitope as the 9F3 antibody;
    16. (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    17. (q) the 9F3 antibody;
    18. (r) an antibody that binds the same epitope as the 24F12 antibody;
    19. (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    20. (t) the 24F 12 antibody;
    21. (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    22. (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    23. (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    24. (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    25. (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the antibody binds the same epitope as the MORAb-003 antibody. In another embodiment, the antibody includes SEQ ID NO: 1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. In another embodiment, the antibody is the MOV18 antibody. In yet another embodiment, the antibody binds the same epitope as the MOV18 antibody. In a further embodiment, the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16). Alternatively or in combination, the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). In certain embodiments, the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the level of FRα in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • In certain embodiments, the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody. Alternatively, or in combination, the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • In certain embodiments, the level of FRα is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • In various embodiments of the foregoing aspects of the invention, the control sample is a standardized control level of FRα in a healthy subject. In another embodiment, the control sample is a sample previously obtained from the subject.
  • In certain embodiments, the sample is treated with guanidine prior to determining the level of FRα in the sample. Alternatively or in combination, the sample is diluted prior to determining the level of FRα in the sample. Alternatively, or in combination, the sample is centrifuged, vortexed, or both, prior to determining the level of FRα in the sample.
  • Methods of Predicting Whether a Subject Will Respond to MORAb-003 treatment
  • In one aspect, the present invention provides a method for predicting whether a subject suffering from an FRα expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the sample derived from the subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the sample derived from the subject and the level of FRα in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • In one aspect, the present invention provides a method for predicting whether a subject suffering from an FRα expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the urine sample derived from the subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the urine sample derived from the subject and the level of FRα in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • In a further aspect, the present invention provides a method for predicting whether a subject suffering from an FRα expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the serum sample derived from the subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the serum sample derived from the subject and the level of FRα in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • In further embodiments, the FRα-expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer. In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In another embodiment, the FRα-expressing lung cancer is non-small cell lung cancer, such as adenocarcinoma.
  • In a further aspect, the present invention provides methods for predicting whether a subject suffering from ovarian cancer will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FRα which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject will respond to treatment with MORAb-003.
  • In various embodiments of the foregoing aspects of the invention, the presence of FRα in the urine sample at a concentration of greater than about 9500 pg/mL, about 10,000 pg/mL, about 11,000 pg/mL, about 12,000 pg/mL, about 13,000 pg/mL, about 14,000 pg/mL, about 15,000 pg/mL, about 16,000 pg/mL, about 17,000 pg/mL, about 18,000 pg/mL, about 19,000 pg/mL, or about 20,000 pg/mL is an indication that the subject is afflicted with ovarian cancer.
  • In various embodiments of the foregoing aspects of the invention, the level of FRα is determined by contacting the sample with an antibody that binds FRα. For example, the antibody is selected from the group consisting of:
    1. (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    2. (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    3. (c) the MOV18 antibody;
    4. (d) an antibody that binds the same epitope as the MOV18 antibody;
    5. (e) the 548908 antibody;
    6. (f) an antibody that binds the same epitope as the 548908 antibody;
    7. (g) the 6D398 antibody;
    8. (h) an antibody that binds the same epitope as the 6D398 antibody;
    9. (i) an antibody that binds the same epitope as the 26B3 antibody;
    10. (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    11. (k) the 26B3 antibody;
    12. (l) an antibody that binds the same epitope as the 19D4 antibody;
    13. (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    14. (n) the 19D4 antibody;
    15. (o) an antibody that binds the same epitope as the 9F3 antibody;
    16. (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    17. (q) the 9F3 antibody;
    18. (r) an antibody that binds the same epitope as the 24F12 antibody;
    19. (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    20. (t) the 24F12 antibody;
    21. (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    22. (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    23. (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    24. (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    25. (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the antibody binds the same epitope as the MORAb-003 antibody. In another embodiment, the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. In another embodiment, the antibody is the MOV 18 antibody. In yet another embodiment, the antibody binds the same epitope as the MOV18 antibody. In a further embodiment, the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16). Alternatively or in combination, the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). In certain embodiments, the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the level of FRα in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • In certain embodiments, the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody. Alternatively, or in combination, the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • In certain embodiments, the level of FRα is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • In various embodiments of the foregoing aspects of the invention, the control sample is a standardized control level of FRα in a healthy subject.
  • In certain embodiments, the sample is treated with guanidine prior to determining the level of FRα in the sample. Alternatively or in combination, the sample is diluted prior to determining the level of FRα in the sample. Alternatively, or in combination, the sample is centrifuged, vortexed, or both, prior to determining the level of FRα in the sample.
  • In a further aspect, the present invention provides a method for predicting whether a subject suffering from an FRα expressing cancer, such as ovarian cancer or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the sample with the level of FRα in a control sample, wherein a difference between the levels of FRα in the sample derived from the subject and in the control sample is an indication that the subject will respond to treatment with MORAb-003; wherein the level of FRα in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody. For example, the sample may be urine, serum, plasma or ascites.
  • In various embodiments of the foregoing aspects of the invention, the MORAb-003 for treatment is (a) an antibody that comprises the heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the light chain amino acid sequence as set forth in SEQ ID NO:8; (b) an antibody that binds the same epitope as the MORAb-003 antibody; or (c) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • Methods of Treating a Subject Having Ovarian Cancer or Lung Cancer
  • In another aspect, the present invention provides methods of treating a subject having ovarian cancer or lung cancer by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from said subject (for example, urine, serum, plasma or ascites); and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein a difference between the level of FRα in the sample derived from said subject and the level of FRα in the control sample is an indication that the subject is afflicted with ovarian cancer or lung cancer; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • In another aspect, the present invention provides methods of treating a subject having ovarian cancer or lung cancer by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a urine sample derived from said subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein a difference between the level of FRα in the urine sample derived from said subject and the level of FRα in the control sample is an indication that the subject is afflicted with ovarian cancer or lung cancer; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • In another aspect, the present invention provides methods of treating a subject having ovarian cancer or lung cancer by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a serum sample derived from said subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein a difference between the level of FRα in the serum sample derived from said subject and the level of FRα in the control sample is an indication that the subject is afflicted with ovarian cancer or lung cancer; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • In a further aspect, the present invention provides methods for treating a subject suffering from ovarian cancer by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a urine sample derived from the subject, wherein the presence of FRα which is not bound to a cell in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject will respond to treatment with MORAb-003; and administering a therapeutically effective amount of MORAb-003 to said subject.
  • In particular embodiments, the level of FRα which is not bound to a cell in the sample derived from said subject is assessed by contacting the sample with an antibody that binds FRα.
  • In various embodiments of the foregoing aspects of the invention, the presence of FRα in the urine sample at a concentration of greater than about 9500 pg/mL, about 10,000 pg/mL, about 11,000 pg/mL, about 12,000 pg/mL, about 13,000 pg/mL, about 14,000 pg/mL, about 15,000 pg/mL, about 16,000 pg/mL, about 17,000 pg/mL, about 18,000 pg/mL, about 19,000 pg/mL, or about 20,000 pg/mL is an indication that the subject is afflicted with ovarian cancer.
  • In various embodiments of the foregoing aspects of the invention, the level of FRα is determined by contacting the sample with an antibody that binds FRα. For example, the antibody is selected from the group consisting of:
    1. (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    2. (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    3. (c) the MOV18 antibody;
    4. (d) an antibody that binds the same epitope as the MOV18 antibody;
    5. (e) the 548908 antibody;
    6. (f) an antibody that binds the same epitope as the 548908 antibody;
    7. (g) the 6D398 antibody;
    8. (h) an antibody that binds the same epitope as the 6D398 antibody;
    9. (i) an antibody that binds the same epitope as the 26B3 antibody;
    10. (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    11. (k) the 26B3 antibody;
    12. (l) an antibody that binds the same epitope as the 19D4 antibody;
    13. (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    14. (n) the 19D4 antibody;
    15. (o) an antibody that binds the same epitope as the 9F3 antibody;
    16. (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    17. (q) the 9F3 antibody;
    18. (r) an antibody that binds the same epitope as the 24F12 antibody;
    19. (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    20. (t) the 24F12 antibody;
    21. (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    22. (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    23. (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    24. (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    25. (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the antibody binds the same epitope as the MORAb-003 antibody. In another embodiment, the antibody includes SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. In another embodiment, the antibody is the MOV 18 antibody. In yet another embodiment, the antibody binds the same epitope as the MOV18 antibody. In a further embodiment, the antibody comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16). Alternatively or in combination, the antibody includes a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). In certain embodiments, the antibody includes (i) the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); or the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In a particular embodiment, the level of FRα in the sample derived from said subject is assessed by contacting the sample with a pair of antibodies selected from the group consisting of (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody; (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody; (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody; and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  • In certain embodiments, the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody. Alternatively, or in combination, the antibody is labeled, for example, with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • In certain embodiments, the level of FRα is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  • In various embodiments of the foregoing aspects of the invention, the control sample is a standardized control level of FRα in a healthy subject.
  • In certain embodiments, the sample is treated with guanidine prior to determining the level of FRα in the sample. Alternatively or in combination, the sample is diluted prior to determining the level of FRα in the sample. Alternatively, or in combination, the sample is centrifuged, vortexed, or both, prior to determining the level of FRα in the sample.
  • In a further aspect, the present invention provides a method for treating a subject suffering from ovarian cancer or lung cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the sample with the level of FRα in a control sample, wherein a difference between the levels of FRα in the sample derived from the subject and in the control sample is an indication that the subject will respond to treatment with MORAb-003; wherein the level of FRα in the sample derived from the subject is assessed by contacting the sample with (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody, (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody, (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody. For example, the sample may be urine, serum, plasma or ascites.
  • In various embodiments of the foregoing aspects of the invention, the MORAb-003 for treatment is (a) an antibody that comprises the heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the light chain amino acid sequence as set forth in SEQ ID NO:8; (b) an antibody that binds the same epitope as the MORAb-003 antibody; or (c) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • Kits of the Invention
  • In one aspect, the present invention provides a kit for assessing whether a subject is afflicted with an FRα-expressing cancer or for assessing the progression of an FRα-expressing cancer in a subject, the kit including means for determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from the subject; and instructions for use of the kit to assess whether the subject is afflicted with an FRα-expressing cancer or to assess the progression of an FRα-expressing cancer. For example, the FRα-expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer. In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In yet another embodiment, the FRα-expressing cancer is non-small cell lung cancer, for example, adenocarcinoma. In a further embodiment, the sample is either urine, serum, plasma or ascites.
  • In a further embodiment, the means includes a folate receptor alpha (FRα) binding agent, for example, an antibody. In a further embodiment, the antibody is selected from the group consisting of
    1. (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    2. (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    3. (c) the MOV18 antibody;
    4. (d) an antibody that binds the same epitope as the MOV18 antibody;
    5. (e) the 548908 antibody;
    6. (f) an antibody that binds the same epitope as the 548908 antibody;
    7. (g) the 6D398 antibody;
    8. (h) an antibody that binds the same epitope as the 6D398 antibody;
    9. (i) an antibody that binds the same epitope as the 26B3 antibody;
    10. (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    11. (k) the 26B3 antibody;
    12. (l) an antibody that binds the same epitope as the 19D4 antibody;
    13. (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    14. (n) the 19D4 antibody;
    15. (o) an antibody that binds the same epitope as the 9F3 antibody;
    16. (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    17. (q) the 9F3 antibody;
    18. (r) an antibody that binds the same epitope as the 24F12 antibody;
    19. (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    20. (t) the 24F12 antibody;
    21. (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    22. (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    23. (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    24. (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    25. (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In certain embodiments, the antibody is labeled including, but not limited to, a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, or an enzyme-label.
  • In yet another embodiment, the kit includes a means for obtaining a sample from the subject.
  • The present invention is further illustrated by the following detailed description and drawings.
  • BRIEF DESCRIPTION OF THE DRAWINGS
    • Figure 1 is a schematic depiction of an electrochemiluminescence immunoassay (ECLIA) method for assessing the FRα in urine as described in the Examples. MOV18 antibody attached to solid supports bound FRα in urine. The FRα was subsequently detected by binding to Ru-labeled MORAb-003 antibody.
    • Figure 2 shows the distribution of FRα levels in the urine of ovarian cancer subjects and normal control subjects as measured by ECLIA (see Example 1).
    • Figure 3 depicts the detection of FRα in urine of ovarian cancer patients (pale band on lane 1, clear band on lane 2) using immunoblotting (see Example 5).
    • Figure 4 shows the distribution of FRα levels in the urine of ovarian cancer subjects and normal control subjects as measured by ECLIA after the urine was treated with guanidine, as described in Example 7.
    • Figure 5 depicts an ROC curve showing the sensitivity and specificity of the ECLIA measurement of FRα levels in urine after the urine was treated with guanidine, as described in Example 7. The area under the curve (AUC) measures the accuracy of the test in separating ovarian cancer from control subjects. A cutoff value (above which the test results were considered abnormal) of 9100 pg/mL was used.
    • Figure 6 shows the distribution of FRα levels in ovarian cancer (OC) and normal control subjects after correction for creatinine levels. There is a statistically significant difference between ovarian cancer patients and controls in creatinine-corrected levels of FRα (p=0.007) (see Example 8).
    • Figure 7 depicts an ROC analysis of creatinine-corrected FRα levels determined using electrochemiluminescence assay (ECLIA) of guanidine-treated urine samples (see Example 8).
    • Figure 8 is a schematic depiction of the enzyme immunoassay (EIA) method used for assessing the level of FRα (i.e., FRα) in samples, as described in Example 9. MOV-18 served as the capture antibody, which bound FRα from biological fluids. The FRα was detected by binding to biotinylated MORAb-003, which was detected using avidin conjugated to horseradish peroxidase (avidin-HRP).
    • Figure 9 depicts results obtained for the measurement of FRα in serum using one- and two-step incubation procedures, as described in Example 9.
    • Figure 10 is a schematic depiction of the three different combinations of capture and detector antibodies that were used with the enzyme immunoassay (EIA) method for assessing the level of FRα in human plasma, as described in Example 11.
    • Figure 11 shows the plasma concentrations of FRα (pg/mL) for individual subjects determined using EIA with three combinations of capture and detector antibodies, as described in Example 11.
    • Figure 12 depicts the relationship between OD values and FRα concentrations (see Example 11).
    • Figure 13 shows the distribution of plasma FRα concentrations in subjects with ovarian cancer and normal control subjects as determined using EIA (see Example 12).
    • Figure 14 depicts the correlation between FRα plasma concentrations determined using EIA and ECLIA (see Example 12).
    • Figure 15 shows correlations between ECLIA measures of FRα levels in serum and urine. The correlation for lung cancer patients was r=0.24 (upper panel) and the correlation for ovarian cancer patients was r=-0.76 (lower panel) (see Example 13).
    • Figure 16 shows the correlation of serum versus plasma FRα levels for assays conducted using pair 1 (see Example 16).
    • Figure 17 shows the correlation of serum versus plasma FRα levels for assays conducted using pair 2 (see Example 16).
    • Figure 18 shows the correlation of serum FRα levels for assays conducted using pair 1 versus pair 2 (see Example 16).
    • Figure 19 shows the correlation of plasma FRα levels for assays conducted using pair 1 versus pair 2 (see Example 16).
    • Figure 20 shows the intraday correlation of serum FRα levels for assays conducted using pair 2 (Example 16).
    DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is based, at least in part, on the unexpected discovery that folate receptor alpha (FRα), not bound to a cell, is found at elevated levels in the body fluids, for example, urine or serum, of a subject having an FRα-expressing cancer such as lung or ovarian cancer as compared to a control sample. Moreover, the present invention is based, at least in part, on the identification of an immunological assay that exhibits the necessary sensitivity for assessing FRα levels in samples, where prior attempts to do so repeatedly failed. As a result, the present invention provides methods for diagnosing an FRα-expressing cancer by assessing levels of an FRα not bound to a cell in samples derived from the subject. Indeed, the present invention overcomes the challenges observed during prior attempts to develop an FRα based diagnostic assay for FRα-expressing cancers such as ovarian cancer by providing an immunological assay capable of accurately assessing levels of FRα not bound to a cell in samples.
  • Accordingly, methods and kits for assessing whether a subject has or is at risk for developing an FRα-expressing cancer and, further, for assessing the progression of FRα-expressing cancer are provided. In various embodiments, the methods involve the comparison of levels of FRα not bound to a cell in samples, for example, urine and serum, as compared to control levels, in assessing the presence, degree or risk of development of ovarian cancer in the subject. In particular embodiments, the methods involve the use of the MORAb-003 antibody, antibodies that bind the same epitope as the MORAb-003 antibody or antibodies having SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3, in assessing the levels of FRα not bound to a cell in a sample, e.g., urine or serum. Alternatively or in addition, the MOV18 antibody or an antibody that binds the same epitope of the MOV18 antibody, the 548908 antibody, an antibody that binds the same epitope of the 548908 antibody, the 6D398 antibody or an antibody that binds the same epitope of the 548908 antibody may be used in accordance with the methods of the present invention.
  • Various aspects of the invention are described in further detail in the following subsections:
  • I. Definitions
  • As used herein, each of the following terms has the meaning associated with it in this section.
  • The articles "a" and "an" are used herein to refer to one or to more than one (i.e. to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.
  • As used herein, the term "subject" refer to human and non-human animals, including veterinary subjects. The term "non-human animal" includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, mice, rabbits, sheep, dog, cat, horse, cow, chickens, amphibians, and reptiles. In a preferred embodiment, the subject is a human.
  • The terms "cancer" or "tumor" are well known in the art and refer to the presence, e.g., in a subject, of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Cancer cells are often in the form of a tumor, but such cells may exist alone within a subject, or may be non-tumorigenic cancer cells, such as leukemia cells. As used herein, the term "cancer" includes pre-malignant as well as malignant cancers.
  • As used herein, an "FRα-expressing cancer" includes any type of cancer characterized in that the cancer cells express FRα. In particular embodiments, the FRα expressing cancer includes cancerous conditions characterized in that the cancer cells are capable of secreting, shedding, exporting or releasing FRα in such a manner that elevated levels of FRα are detectable in a biological sample from the subject. FRα-expressing cancers include, but are not limited to, lung cancer (e.g., bronchioalveolar carcinomas, carcinoid tumors, and non-small cell lung cancers, such as adenocarcinomas); mesothelioma; ovarian cancer; renal cancer; brain cancer (e.g., anaplastic ependymoma and cerebellar juvenile pilocytic astrocytoma); cervical cancer; nasopharyngeal cancer; mesodermally derived tumor; squamous cell carcinoma of the head and neck; endometrial cancer; endometrioid adenocarcinomas of the ovary, serous cystadenocarcinomas, breast cancer; bladder cancer; pancreatic cancer; bone cancer (e.g., high-grade osteosarcoma); pituitary cancer (e.g., pituitary adenoma). See e.g., U.S. Patent No. 7,754,698 ; U.S. Patent Application No. 2005/0232919 ; WO 2009/132081 ; Bueno R et al. J of Thoracic and Cardiovascular Surgery, 121(2) : 225-233 (2001); Elkanat H & Ratnam M. Frontiers in Bioscience, 11, 506-519 (2006); Franklin, WA et al. Int J Cancer, Suppl 8: 89-95 (1994); Hartman L.C. et al. Int J Cancer 121: 938-942 (2007); Iwakiri S et al. Annals of Surgical Oncology, 15(3): 889-899; Weitman, SD et al. Cancer Res 52: 3396-3401 (1992); Saba N.F. et al. Head Neck, 31(4): 475-481 (2009); Yang R et al. Clin Cancer Res 13: 2557-2567 (2007). In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In another embodiment, the FRα-expressing cancer is lung cancer such as non-small cell lung cancer. In other embodiments, the FRα-expressing cancer is colorectal cancer and medullary thyroid cancer.
  • As used herein, a subject who is "afflicted with an FRα-expressing cancer" is one who is clinically diagnosed with such a cancer by a qualified clinician (for example, by the methods of the present invention), or one who exhibits one or more signs or symptoms (for example, elevated levels of FRα in biological fluids) of such a cancer and is subsequently clinically diagnosed with such a cancer by a qualified clinician (for example, by the methods of the present invention). A non-human subject that serves as an animal model of FRα-expressing cancer may also fall within the scope of the term a subject "afflicted with an FRα-expressing cancer."
  • The term "ovarian cancer" refers to the art recognized disease and includes each of epithelial ovarian cancer (EOC; >90% of ovarian cancer in Western countries), germ cell tumors (circa 2-3% of ovarian cancer), and stromal ovarian cancer. Ovarian cancer is stratified into different groups based on the differentiation of the tumor tissue. In grade I, the tumor tissue is well differentiated. In grade II, tumor tissue is moderately well differentiated. In grade III, the tumor tissue is poorly differentiated. This grade correlates with a less favorable prognosis than grades I and II.
  • Ovarian cancer is stratified into different stages based on the spread of the cancer. Stage I is generally confined within the capsule surrounding one (stage IA) or both (stage IB) ovaries, although in some stage I (i.e. stage IC) cancers, malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries. Stage II involves extension or metastasis of the tumor from one or both ovaries to other pelvic structures. In stage IIA, the tumor extends or has metastasized to the uterus, the fallopian tubes, or both. Stage IIB involves extension of the tumor to the pelvis. Stage IIC is stage IIA or IIB in which malignant cells may be detected in ascites, in peritoneal rinse fluid, or on the surface of the ovaries. In stage III, the tumor comprises at least one malignant extension to the small bowel or the omentum, has formed extrapelvic peritoneal implants of microscopic (stage IIIA) or macroscopic (< 2 centimeter diameter, stage IIIB; > 2 centimeter diameter, stage IIIC) size, or has metastasized to a retroperitoneal or inguinal lymph node (an alternate indicator of stage IIIC). In stage IV, distant (i.e. non-peritoneal) metastases of the tumor can be detected.
  • The durations of the various stages of ovarian cancer are not presently known, but are believed to be at least about a year each ( Richart et al., 1969, Am. J. Obstet. Gynecol. 105:386). Prognosis declines with increasing stage designation. For example, 5-year survival rates for human subjects diagnosed with stage I, II, III, and IV ovarian cancer are 80%, 57%, 25%, and 8%, respectively.
  • Each of the foregoing types, groups and stages of ovarian cancer are encompassed by the term "ovarian cancer" as used herein.
  • As used herein, the term "lung cancer" refers to a disease in tissues of the lung involving uncontrolled cell growth, which, in some cases, leads to metastasis. Lung cancer is the most common cause of cancer-related death in men and women. The majority of primary lung cancers are carcinomas of the lung, derived from epithelial cells. The main types of lung cancer are small cell lung carcinoma (SCLC) and non-small cell lung carcinoma (NSCLC). In a particular embodiment, the FRα-expressing cancer is a non-small cell lung cancer.
  • Small cell lung cancer or small cell lung carcinoma (SCLC) is a malignant cancer of the lung, wherein the cancer cells have a flat shape and scanty cytoplasm; therefore, SCLC is sometimes called "oat cell carcinoma." SCLC is generally more metastatic than NSCLC and is sometimes seen in combination with squamous cell carcinomas.
  • As used herein, the term "non-small cell lung cancer," also known as non-small cell lung carcinoma (NSCLC), refers to epithelial lung cancer other than small cell lung carcinoma (SCLC). There are three main sub-types: adenocarcinoma, squamous cell lung carcinoma, and large cell lung carcinoma. Other less common types of non-small cell lung cancer include pleomorphic, carcinoid tumor, salivary gland carcinoma, and unclassified carcinoma. Adenocarcinomas account for approximately 40% of lung cancers, and are the most common type of lung cancer in people who have never smoked. Squamous cell carcinomas account for about 25% of lung cancers. Squamous cell carcinoma of the lung is more common in men than in women and is even more highly correlated with a history of tobacco smoking than are other types of lung carcinoma. There are at least four variants (papillary, small cell, clear cell, and basaloid) of squamous cell carcinoma of the lung. Large cell lung carcinomas are a heterogeneous group of malignant neoplasms originating from transformed epithelial cells in the lung. Large cell lung carcinomas are carcinomas that lack light microscopic characteristics of small cell carcinoma, squamous cell carcinoma, or adenocarcinoma.
  • Different staging systems are used for SCLC and NSCLC. SCLC is categorized as limited disease confined to the ipsilateral hemithorax or as extensive disease with metastasis beyond the ipsilateral hemithorax.
  • NSCLC may be categorized using the tumor-nodes-metastasis (TNM) staging system. See Spira J & Ettinger, D.S. Multidisciplinary management of lung cancer, N Engl J Med, 350:382- (2004) (hereinafter Spira); Greene FL, Page DL, Fleming ID, Fritz AG, Balch CM, Haller DG, et al (eds). AJCC Cancer Staging Manual. 6th edition. New York: Springer-Verlag, 2002:167-77 (hereinafter Greene); Sobin LH, Wittekind CH (eds). International Union Against Cancer. TNM classification of malignant tumours. 6th edition. New York: Wiley-Liss (2002) (hereinafter Sobin). In addition, NSCLC is typically treated according to the stage of cancer determined by the following classification scheme (see http://www.cancer.gov/cancertopics/pdq/treatment/non-small-cell-lung/Patient/page2#Keypoint10).
  • In the occult (hidden) stage, cancer cells are found in sputum (mucus coughed up from the lungs), but no tumor can be found in the lung by imaging or bronchoscopy, or the tumor is too small to be checked.
  • In stage 0 (carcinoma in situ), abnormal cells are found in the lining of the airways. These abnormal cells may become cancer and spread into nearby normal tissue. Stage 0 is also called carcinoma in situ.
  • Stage I, in which cancer has formed, is divided into stages IA and IB.
  • In Stage IA, the tumor is in the lung only and is 3 centimeters or smaller.
  • In Stage IB, the cancer has not spread to the lymph nodes and one or more of the following is true: (i) The tumor is larger than 3 centimeters but not larger than 5 centimeters; (ii) cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus; (iii) cancer has spread to the innermost layer of the membrane that covers the lung; (iv) part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus.
  • In Stage IIA, cancer has spread to certain lymph nodes on the same side of the chest as the primary tumor; the cancer is (a) 5 cm or smaller, (b) has spread to the main bronchus, and/or (c) has spread to the innermost layer of the lung lining. OR, cancer has not spread to lymph nodes; the cancer is (d) larger than 5 cm but not larger than 7 cm, (e) has spread to the main bronchus, and/or (f) has spread to the innermost layer of the lung lining. Part of the lung may have collapsed or become inflamed. Stage IIA is divided into two sections depending on the size of the tumor, where the tumor is found, and whether there is cancer in the lymph nodes. In the first section, cancer has spread to lymph nodes on the same side of the chest as the tumor. The lymph nodes with cancer are within the lung or near the bronchus. Also, one or more of the following is true: (i) the tumor is not larger than 5 centimeters, (ii) cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus, (iii) cancer has spread to the innermost layer of the membrane that covers the lung, (iv) part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus. In the second section, cancer has not spread to lymph nodes and one or more of the following is true: (i) the tumor is larger than 5 centimeters but not larger than 7 centimeters, (ii) cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus, (iii) cancer has spread to the innermost layer of the membrane that covers the lung, (iv) part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus.
  • In Stage IIB, cancer has spread to certain lymph nodes on the same side of the chest as the primary tumor; the cancer is (a) larger than 5 cm but not larger than 7 cm, (b) has spread to the main bronchus, and/or (c) has spread to the innermost layer of the lung lining. Part of the lung may have collapsed or become inflamed. Alternatively, (d) the cancer is larger than 7 cm; (e) has spread to the main bronchus, (f) the diaphragm, (g) the chest wall or the lining of the chest wall; and/or (h) has spread to the membrane around the heart. There may be one or more separate tumors in the same lobe of the lung; cancer may have spread to the nerve that controls the diaphragm; the whole lung may have collapsed or become inflamed. Stage IIB is divided into two sections depending on the size of the tumor, where the tumor is found, and whether there is cancer in the lymph nodes. In the first section, cancer has spread to nearby lymph nodes on the same side of the chest as the tumor. The lymph nodes with cancer are within the lung or near the bronchus. Also, one or more of the following is true: (i) the tumor is larger than 5 centimeters but not larger than 7 centimeters, (ii) cancer has spread to the main bronchus and is at least 2 centimeters below where the trachea joins the bronchus, (iii) cancer has spread to the innermost layer of the membrane that covers the lung, (iv) part of the lung has collapsed or developed pneumonitis (inflammation of the lung) in the area where the trachea joins the bronchus. In the second section, cancer has not spread to lymph nodes and one or more of the following is true: (i) the tumor is larger than 7 centimeters, (ii) cancer has spread to the main bronchus (and is less than 2 centimeters below where the trachea joins the bronchus), the chest wall, the diaphragm, or the nerve that controls the diaphragm, (iii) cancer has spread to the membrane around the heart or lining the chest wall, (iv) the whole lung has collapsed or developed pneumonitis (inflammation of the lung), (v) there are one or more separate tumors in the same lobe of the lung.
  • Stage IIIA is divided into three sections depending on the size of the tumor, where the tumor is found, and which lymph nodes have cancer (if any). In the first section of Stage IIIA, cancer has spread to lymph nodes on the same side of the chest as the tumor. The lymph nodes with cancer are near the sternum (chest bone) or where the bronchus enters the lung. Also, the tumor may be any size; part of the lung (where the trachea joins the bronchus) or the whole lung may have collapsed or developed pneumonitis (inflammation of the lung); there may be one or more separate tumors in the same lobe of the lung; and cancer may have spread to any of the following: (i) main bronchus, but not the area where the trachea joins the bronchus, (ii) chest wall, (iii) diaphragm and the nerve that controls it, (iv) membrane around the lung or lining the chest wall, (iv) membrane around the heart. In the second section of Stage IIIA, cancer has spread to lymph nodes on the same side of the chest as the tumor. The lymph nodes with cancer are within the lung or near the bronchus. Also, the tumor may be any size; the whole lung may have collapsed or developed pneumonitis (inflammation of the lung); there may be one or more separate tumors in any of the lobes of the lung with cancer; and cancer may have spread to any of the following: (i) main bronchus, but not the area where the trachea joins the bronchus, (ii) chest wall, (iii) diaphragm and the nerve that controls it, (iv) membrane around the lung or lining the chest wall, (v) heart or the membrane around it, (vi) major blood vessels that lead to or from the heart, (vi) trachea, (vii) esophagus, (viii) nerve that controls the larynx (voice box), (ix) sternum (chest bone) or backbone, (x) carina (where the trachea joins the bronchi). In the third section of Stage IIIA, cancer has not spread to the lymph nodes and the tumor may be any size, and cancer has spread to any of the following: (i) heart, (ii) major blood vessels that lead to or from the heart, (iii) trachea, (iv) esophagus, (v) nerve that controls the larynx (voice box), (vi) sternum (chest bone) or backbone, (vi) carina (where the trachea joins the bronchi).
  • Stage IIIB is divided into two sections depending on the size of the tumor, where the tumor is found, and which lymph nodes have cancer. In the first section, cancer has spread to lymph nodes above the collarbone or to lymph nodes on the opposite side of the chest as the tumor; the tumor may be any size; part of the lung (where the trachea joins the bronchus) or the whole lung may have collapsed or developed pneumonitis (inflammation of the lung); there may be one or more separate tumors in any of the lobes of the lung with cancer; and cancer may have spread to any of the following: (i) main bronchus, (ii) chest wall, (iii) diaphragm and the nerve that controls it, (iv) membrane around the lung or lining the chest wall, (iv) heart or the membrane around it, (v) major blood vessels that lead to or from the heart, (vi) trachea, (vii) esophagus, (viii) nerve that controls the larynx (voice box), (ix) sternum (chest bone) or backbone, (x) carina (where the trachea joins the bronchi). In the second section of Stage IIIB, cancer has spread to lymph nodes on the same side of the chest as the tumor; the lymph nodes with cancer are near the sternum (chest bone) or where the bronchus enters the lung; the tumor may be any size; there may be separate tumors in different lobes of the same lung; and cancer has spread to any of the following: (i) heart, (ii) major blood vessels that lead to or from the heart, (iii) trachea, (iv) esophagus, (v) nerve that controls the larynx (voice box), (vi) sternum (chest bone) or backbone, (vii) carina (where the trachea joins the bronchi).
  • In Stage IV, the tumor may be any size and cancer may have spread to lymph nodes. One or more of the following is true: there are one or more tumors in both lungs; cancer is found in fluid around the lungs or the heart; cancer has spread to other parts of the body, such as the brain, liver, adrenal glands, kidneys, or bone.
  • Accordingly, in various embodiments of the foregoing invention, the lung cancer may be stratified into any of the preceding stages (e.g., occult, stage 0, stage IA, stage IB, stage IIA, stage IIB, stage IIIA, stage IIIB or stage IV) based on assessing of the levels of FRα not bound to a cell, such as a normal or cancerous cell, in a sample (for example, urine or serum) of a subject.
  • As used herein, the term "folate receptor alpha" (also referred to as FRα, FR-alpha, FOLR-1 or FOLR1) refers to the alpha isoform of the high affinity receptor for folate. Membrane bound FRα is attached to the cell surface by a glycosyl phosphatidylinositol (GPI) anchor), recycles between extracellular and endocytic compartments and is capable of transporting folate into the cell. FRα is expressed in a variety of epithelial tissues including those of the female reproductive tract, placenta, breast, kidney proximal tubules, choroid plexus, lung and salivary glands. Soluble forms of FRα may be derived by the action of proteases or phospholipase on membrane anchored folate receptors.
  • The consensus nucleotide and amino acid sequences for human FRα are set forth herein as SEQ ID NOs: 24 and 25, respectively. Variants, for example, naturally occurring allelic variants or sequences containing at least one amino acid substitution, are encompassed by the terms as used herein.
  • As used herein, the term "not bound to a cell" refers to FRα that is not attached to the cellular membrane of a cell, such as a cancerous cell. In a particular embodiment, the FRα not bound to a cell is unbound to any cell and is freely floating or solubilized in biological fluids, e.g., urine or serum. For example, the FRα may be shed, secreted or exported from normal or cancerous cells, for example, from the surface of cancerous cells, into biological fluids.
  • The "level" of folate receptor alpha not bound to a cell, as used herein, refers to the level of folate receptor alpha protein as determined using any method known in the art for the measurement of protein levels. Such methods include, for example, electrophoresis, capillary electrophoresis, high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, fluid or gel precipitin reactions, absorption spectroscopy, a colorimetric assays, spectrophotometric assays, flow cytometry, immunodiffusion (single or double), solution phase assay, immunoelectrophoresis, Western blotting, radioimmunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, and electrochemiluminescence immunoassay (exemplified below), and the like. In a preferred embodiment, the level is determined using antibody-based techniques, as described in more detail herein.
  • It is generally preferable to immobilize either an antibody or binding protein specific for FRα not bound to a cell on a solid support for Western blots and immunofluorescence techniques. Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • One skilled in the art will know many other suitable carriers for binding antibody or antigen, and will be able to adapt such support for use with the present invention. For example, protein isolated from a subject sample (e.g., urine or serum) can be run on a polyacrylamide gel electrophoresis and immobilized onto a solid phase support such as nitrocellulose. The support can then be washed with suitable buffers followed by treatment with the labeled antibody. The solid phase support can then be washed with the buffer a second time to remove unbound antibody. The amount of bound label on the solid support can then be detected by conventional means. Means of detecting proteins using electrophoretic techniques are well known to those of skill in the art (see generally, R. Scopes (1982) Protein Purification, Springer-Verlag, N.Y.; Deutscher, (1990) Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc., N.Y.).
  • Other standard methods include immunoassay techniques which are well known to one of ordinary skill in the art and may be found in Principles And Practice Of Immunoassay, 2nd Edition, Price and Newman, eds., MacMillan (1997) and Antibodies, A Laboratory Manual, Harlow and Lane, eds., Cold Spring Harbor Laboratory, Ch. 9 (1988), each of which is incorporated herein by reference in its entirety.
  • Antibodies used in immunoassays to determine the level of expression of folate receptor alpha may be labeled with a detectable label. The term "labeled", with regard to the binding agent or antibody, is intended to encompass direct labeling of the binding agent or antibody by coupling (i.e., physically linking) a detectable substance to the binding agent or antibody, as well as indirect labeling of the binding agent or antibody by reactivity with another reagent that is directly labeled. An example of indirect labeling includes detection of a primary antibody using a fluorescently labeled secondary antibody. In one embodiment, the antibody is labeled, e.g., radio-labeled, chromophore-labeled, fluorophore-labeled, or enzyme-labeled. In another embodiment, the antibody is an antibody derivative (e.g., an antibody conjugated with a substrate or with the protein or ligand of a protein-ligand pair (e.g., biotin-streptavidin), or an antibody fragment (e.g., a single-chain antibody, an isolated antibody hypervariable domain) which binds specifically with FRα not bound to a cell.
  • In one embodiment of the invention, proteomic methods, e.g., mass spectrometry, are used. Mass spectrometry is an analytical technique that consists of ionizing chemical compounds to generate charged molecules (or fragments thereof) and measuring their mass-to-charge ratios. In a typical mass spectrometry procedure, a sample is obtained from a subject, loaded onto the mass spectrometry, and its components (e.g., FRα) are ionized by different methods (e.g., by impacting them with an electron beam), resulting in the formation of charged particles (ions). The mass-to-charge ratio of the particles is then calculated from the motion of the ions as they transit through electromagnetic fields.
  • For example, matrix-associated laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) or surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF MS) which involves the application of a sample, such as urine or serum, to a protein-binding chip (Wright, G.L., Jr., et al. (2002) Expert Rev Mol Diagn 2:549; Li, J., et al. (2002) Clin Chem 48:1296; Laronga, C., et al. (2003) Dis Markers 19:229; Petricoin, E.F., et al. (2002) 359:572; Adam, B.L., et al. (2002) Cancer Res 62:3609; Tolson, J., et al. (2004) Lab Invest 84:845; Xiao, Z., et al. (2001) Cancer Res 61:6029) can be used to determine the level of FRα.
  • Furthermore, in vivo techniques for determination of the level of FRα not bound to a cell include introducing into a subject a labeled antibody directed against FRα, which binds to and transforms FRα into a detectable molecule. The presence, level, or location of the detectable FRα not bound to a cell in a subject may be determined using standard imaging techniques.
  • The term "sample" as used herein refers to a collection of similar fluids, cells, or tissues isolated from a subject, as well as fluids, cells, or tissues present within a subject. In preferred embodiments the sample is a biological fluid containing FRα not bound to a cancerous cell. Biological fluids are typically liquids at physiological temperatures and may include naturally occurring fluids present in, withdrawn from, expressed or otherwise extracted from a subject or biological source. Certain biological fluids derive from particular tissues, organs or localized regions and certain other biological fluids may be more globally or systemically situated in a subject or biological source. Examples of biological fluids include blood, serum and serosal fluids, plasma, lymph, urine, cerebrospinal fluid, saliva, ocular fluids, cystic fluid, tear drops, feces, sputum, mucosal secretions of the secretory tissues and organs, vaginal secretions, gynecological fluids, ascites fluids such as those associated with non-solid tumors, fluids of the pleural, pericardial, peritoneal, abdominal and other body cavities, fluids collected by bronchial lavage and the like. In a particular embodiment, the sample is urine or serum. In another embodiment, the sample does not include ascites or is not an ascite sample. In another embodiment, the sample does not include peritoneal fluid or is not peritoneal fluid.
  • In one embodiment, the sample is removed from the subject. In another embodiment, the sample is present within the subject. Biological fluids may also include liquid solutions contacted with a subject or biological source, for example, cell and organ culture medium including cell or organ conditioned medium, lavage fluids and the like.
  • In some embodiments, only a portion of the sample is subjected to an assay for determining the level of FRα not bound to a cell, or various portions of the sample are subjected to various assays for determining the level of FRα not bound to a cell. Also, in many embodiments, the sample may be pre-treated by physical or chemical means prior to the assay. For example, in embodiments discussed in more detail in the Examples section, samples, for example, urine samples, were subjected to centrifugation, dilution and/or treatment with a solubilizing substance (e.g., guanidine treatment) prior to assaying the samples for FRα not bound to a cell. Such techniques serve to enhance the accuracy, reliability and reproducibility of the assays of the present invention.
  • The term "control sample," as used herein, refers to any clinically relevant control sample, including, for example, a sample from a healthy subject not afflicted with ovarian cancer, a sample from a subject having a less severe or slower progressing ovarian cancer than the subject to be assessed, a sample from a subject having some other type of cancer or disease, and the like. A control sample may include a sample derived from one or more subjects. A control sample may also be a sample made at an earlier timepoint from the subject to be assessed. For example, the control sample could be a sample taken from the subject to be assessed before the onset of the FRα expressing cancer such as lung or ovarian cancer, at an earlier stage of disease, or before the administration of treatment or of a portion of treatment. The control sample may also be a sample from an animal model, or from a tissue or cell lines derived from the animal model, of the FRα expressing cancer such as lung or ovarian cancer. The level of FRα not bound to a cell in a control sample that consists of a group of measurements may be determined based on any appropriate statistical measure, such as, for example, measures of central tendency including average, median, or modal values.
  • The term "control level" refers to an accepted or pre-determined level of FRα which is used to compare with the level of FRα in a sample derived from a subject. In one embodiment, the control level of FRα is based on the level of FRα not bound to a cell in sample(s) from a subject(s) having slow disease progression. In another embodiment, the control level of FRα not bound to a cell is based on the level in a sample from a subject(s) having rapid disease progression. In another embodiment, the control level of FRα is based on the level of FRα not bound to a cell in a sample(s) from an unaffected, i.e., non-diseased, subject(s), i.e., a subject who does not have an FRα expressing cancer such as lung or ovarian cancer. In yet another embodiment, the control level of FRα is based on the level of FRα not bound to a cell in a sample from a subject(s) prior to the administration of a therapy for ovarian cancer. In another embodiment, the control level of FRα is based on the level of FRα not bound to a cell in a sample(s) from a subject(s) having an FRα expressing cancer such as lung or ovarian cancer that is not contacted with a test compound. In another embodiment, the control level of FRα is based on the level of FRα not bound to a cell in a sample(s) from a subject(s) not having an FRα expressing cancer such as lung or ovarian cancer that is contacted with a test compound. In one embodiment, the control level of FRα is based on the level of FRα not bound to a cell in a sample(s) from an animal model of an FRα expressing cancer such as lung or ovarian cancer, a cell, or a cell line derived from the animal model of an FRα expressing cancer such as lung or ovarian cancer.
  • In one embodiment, the control is a standardized control, such as, for example, a control which is predetermined using an average of the levels of FRα not bound to a cell from a population of subjects having no FRα expressing cancer such as lung or ovarian cancer. In still other embodiments of the invention, a control level of FRα is based on the level of FRα not bound to a cell in a non-cancerous sample(s) derived from the subject having an FRα expressing cancer such as lung or ovarian cancer. For example, when a laparotomy or other medical procedure reveals the presence of ovarian cancer in one portion of the ovaries, the control level of FRα may be determined using the non-affected portion of the ovaries, and this control level may be compared with the level of FRα in an affected portion of the ovaries. Similarly, when a biopsy or other medical procedure reveals the presence of a lung cancer in one portion of the lungs, the control level of FRα may be determined using the non-affected portion of the lungs, and this control level may be compared with the level of FRα in an affected portion of the lungs.
  • As used herein, "a difference" between the level of folate receptor alpha not bound to a cell in a sample from a subject (i.e., a test sample) and the level of folate receptor alpha not bound to a cell in a control sample refers broadly to any clinically relevant and/or statistically significant difference in the level of folate receptor alpha in the two samples. In an exemplary embodiment, the difference is selected based on a cutoff value determined using a receiver operating characteristic (ROC) analysis, an example of which is presented in Example 6.
  • In other embodiments, the difference must be greater than the limits of detection of the method for determining the level of FRα not bound to a cell. It is preferred that the difference be at least greater than the standard error of the assessment method, and preferably a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method. The difference may be assessed by any appropriate comparison, including any appropriate parametric or nonparametric descriptive statistic or comparison. For example, "an increase" in the level of FRα not bound to a cell may refer to a level in a test sample that is about two, and more preferably about three, about four, about five, about six, about seven, about eight, about nine, about ten or more times more than the level of FRα in the control sample. An increase may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations above the average level of FRα in the control sample. Likewise, "a decrease" in the level of FRα not bound to a cell may refer to a level in a test sample that is preferably at least about two, and more preferably about three, about four, about five, about six, about seven, about eight, about nine, about ten or more times less than the level of FRα in the control sample. A decrease may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations below the average level of FRα in the control sample.
  • As used herein, the term "contacting the sample" with an FRα binding agent, e.g., an anti-FRa antibody, includes exposing the sample, or any portion thereof with the agent or antibody, such that at least a portion of the sample comes into contact with the agent or antibody. The sample or portion thereof may be altered in some way, such as by subjecting it to physical or chemical treatments (e.g., dilution or guanidine treatment), prior to the act of contacting it with the agent or antibody.
  • The term "antibody" as used herein, comprises four polypeptide chains, two heavy (H) chains and two light (L) chains, interconnected by disulfide bonds, as well as any functional (i.e., antigen-binding) fragment, mutant, variant, or derivation thereof, which retains the essential epitope binding features of an Ig molecule. Such mutant, variant, or derivative antibody formats are known in the art, and include molecules such as Fab fragments, Fab' fragments, F(ab')2 fragments, Fd fragments, Fabc fragments, Sc antibodies (single chain antibodies), diabodies, individual antibody light chains, individual antibody heavy chains, chimeric fusions between antibody chains and the like. Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively.
  • The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • The term "antigen-binding portion" of an antibody, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., FRα not bound to a cell). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb including VH and VL domains; (vi) a dAb fragment (Ward et al. (1989) Nature 341, 544-546), which consists of a VH domain; (vii) a dAb which consists of a VH or a VL domain; and (viii) an isolated complementarity determining region (CDR) or (ix) a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242, 423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85, 5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins.
  • The term "antibody", as used herein, includes polyclonal antibodies, monoclonal antibodies, murine antibodies, chimeric antibodies, humanized antibodies, and human antibodies, and those that occur naturally or are recombinantly produced according to methods well known in the art.
  • In one embodiment, an antibody for use in the methods of the invention is a bispecific antibody. A "bispecific antibody" is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, (1990) Clin. Exp. Immunol. 79, 315-321; Kostelny et al. (1992) J. Immunol. 148, 1547-1553.
  • In another embodiment, an antibody for use in the methods of the invention is a camelid antibody as described in, for example, PCT Publication WO 94/04678 , the entire contents of which are incorporated herein by reference.
  • A region of the camelid antibody that is the small, single variable domain identified as VHH can be obtained by genetic engineering to yield a small protein having high affinity for a target, resulting in a low molecular weight, antibody-derived protein known as a "camelid nanobody". See U.S. Pat. No. 5,759,808 ; see also Stijlemans et al., 2004 J. Biol. Chem. 279: 1256-1261; Dumoulin et al., 2003 Nature 424: 783-788; Pleschberger et al., 2003 Bioconjugate Chem. 14: 440-448; Cortez-Retamozo et al., 2002 Int. J. Cancer 89: 456-62; and Lauwereys et al., 1998 EMBO J. 17: 3512-3520. Engineered libraries of camelid antibodies and antibody fragments are commercially available, for example, from Ablynx, Ghent, Belgium. Accordingly, a feature of the present invention is a camelid nanobody having high affinity for FRα.
  • In other embodiments of the invention, an antibody for use in the methods of the invention is a diabody, a single chain diabody, or a di-diabody.
  • Diabodies are bivalent, bispecific molecules in which VH and VL domains are expressed on a single polypeptide chain, connected by a linker that is too short to allow for pairing between the two domains on the same chain. The VH and VL domains pair with complementary domains of another chain, thereby creating two antigen binding sites (see e.g., Holliger et al., 1993 Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak et al., 1994 Structure 2:1121-1123). Diabodies can be produced by expressing two polypeptide chains with either the structure VHA-VLB and VHB-VLA (VH-VL configuration), or VLA-VHB and VLB-VHA (VL-VH configuration) within the same cell. Most of them can be expressed in soluble form in bacteria.
  • Single chain diabodies (scDb) are produced by connecting the two diabody-forming polypeptide chains with linker of approximately 15 amino acid residues (see Holliger and Winter, 1997 Cancer Immunol. Immunother., 45(3-4):128-30; Wu et al., 1996 Immunotechnology, 2(1):21-36). scDb can be expressed in bacteria in soluble, active monomeric form (see Holliger and Winter, 1997 Cancer Immunol. Immunother., 45(34): 128-30; Wu et al., 1996 Immunotechnology, 2(1):21-36; Pluckthun and Pack, 1997 Immunotechnology, 3(2): 83-105; Ridgway et al., 1996 Protein Eng., 9(7):617-21).
  • A diabody can be fused to Fc to generate a "di-diabody" (see Lu et al., 2004 J. Biol. Chem., 279(4):2856-65).
  • FRα binding molecules that exhibit functional properties of antibodies but derive their framework and antigen binding portions from other polypeptides (e.g., polypeptides other than those encoded by antibody genes or generated by the recombination of antibody genes in vivo) may also be used in the methods of the present invention. The antigen binding domains (e.g., FRα binding domains) of these binding molecules are generated through a directed evolution process. See U.S. Pat. No. 7,115,396 . Molecules that have an overall fold similar to that of a variable domain of an antibody (an "immunoglobulin-like" fold) are appropriate scaffold proteins. Scaffold proteins suitable for deriving antigen binding molecules include fibronectin or a fibronectin dimer, tenascin, N-cadherin, E-cadherin, ICAM, titin, GCSF-receptor, cytokine receptor, glycosidase inhibitor, antibiotic chromoprotein, myelin membrane adhesion molecule P0, CD8, CD4, CD2, class I MHC, T-cell antigen receptor, CD1, C2 and I-set domains of VCAM-1, I-set immunoglobulin domain of myosin-binding protein C, I-set immunoglobulin domain of myosin-binding protein H, I-set immunoglobulin domain of telokin, NCAM, twitchin, neuroglian, growth hormone receptor, erythropoietin receptor, prolactin receptor, interferon-gamma receptor, P-galactosidase/glucuronidase, β-glucuronidase, transglutaminase, T-cell antigen receptor, superoxide dismutase, tissue factor domain, cytochrome F, green fluorescent protein, GroEL, and thaumatin.
  • "Specific binding" when used in the context of antibodies, or antibody fragments, represents binding via domains encoded by immunoglobulin genes or fragments of immunoglobulin genes to one or more epitopes of a protein of interest, but which do not substantially recognize and bind other molecules in a sample containing a mixed population of antigenic molecules. Typically, an antibody binds to a cognate antigen with a Kd of less than about 1x10-8 M, as measured by a surface plasmon resonance assay or a cell binding assay.
  • As used herein, a folate receptor alpha "binding agent" includes an antibody that binds FRα not bound to a cell as well as non-antibody binding agents. To generate non-antibody binding agents or binding molecules, a library of clones can be created in which sequences in regions of the scaffold protein that form antigen binding surfaces (e.g., regions analogous in position and structure to CDRs of an antibody variable domain immunoglobulin fold) are randomized. Library clones are tested for specific binding to the antigen of interest (e.g., FRα) and for other functions (e.g., inhibition of biological activity of FRα). Selected clones can be used as the basis for further randomization and selection to produce derivatives of higher affinity for the antigen.
  • High affinity binding molecules are generated, for example, using the tenth module of fibronectin III (10Fn3) as the scaffold, described in U.S. Pat. Nos. 6,818,418 and 7,115,396 ; Roberts and Szostak, 1997 Proc. Natl. Acad. Sci USA 94:12297; U.S. Pat. No. 6,261,804 ; U.S. Pat. No. 6,258,558 ; and Szostak et al. WO98/31700 , the entire contents of each of which are incorporated herein by reference.
  • Non-antibody binding molecules can be produced as dimers or multimers to increase avidity for the target antigen. For example, the antigen binding domain is expressed as a fusion with a constant region (Fc) of an antibody that forms Fc-Fc dimers. See, e.g., U.S. Pat. No. 7,115,396 , the entire contents of which are incorporated herein by reference.
  • An "antigen" is a molecule recognized by the immune system; the term originally came from "antibody generator" and includes a molecule that binds specifically to an antibody. At the molecular level, an antigen is characterized by its ability to be bound at the antigen-binding site of an antibody. In the present invention, the antigen is FRα, such as FRα which is not bound to a cell FRα or a portion thereof.
  • As used herein, the term "epitope" refers to the molecular surface features of an antigen, e.g., FRα, capable of being bound by an antibody. Antigenic molecules, normally being "large" biological polymers, usually present several surface features that can act as points of interaction for specific antibodies. Any such distinct molecular feature constitutes an epitope. Most antigens therefore have the potential to be bound by several distinct antibodies, each of which is typically specific to a particular epitope. In one embodiment of the present invention, a binding agent, e.g., antibody, binds to an epitope on FRα which is available in the form of the receptor which is not bound to a cell but not in the membrane bound form of the receptor. For example, the antibody may bind to the same epitope on FRα to which MORAB-003 binds.
  • As used herein, the phrase "progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer" includes the progression of such a cancer from a less severe to a more severe state. This could include an increase in the number or severity of tumors, the degree of metastasis, the speed with which the cancer is growing and spreading, and the like. For example, "the progression of ovarian cancer" includes the progression of such a cancer from a less severe to a more severe state, such as the progression from Stage I to Stage II, from Stage II to Stage III, etc. Alternatively, the phrase "progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer" may refer to the regression of an FRα-expressing cancer from a more severe state to a less severe state. For example, in one embodiment, "the progression of ovarian cancer" refers to the regression from Stage IV to Stage III, from Stage III to Stage II, etc. In other embodiments, the "progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer" may refer to the survival rate determined from the beginning of symptoms of the FRα-expressing cancer, or to the survival rate from the time of diagnosis of the FRα-expressing cancer.
  • As used herein, the term "stratifying" refers to characterizing an FRα expressing cancer, for example, ovarian or lung cancer, into an appropriate stage based, for example, on the degree of the spread of the cancer, as well accepted stratifications in the art. For example, stratifying includes characterizing the FRα expressing cancer into Stage I, Stage II, Stage III or Stage IV. In certain embodiments, Stage I refers to cancers that are localized to one part of the body. In certain embodiments, Stages II and III refer to cancers that are locally advanced, wherein a distinction between the stages are often specific to the particular cancer. Finally, Stage IV refers to cancers that have often metastasized, or spread to other organs or throughout the body.
  • As used herein, the term "survival" refers to the continuation of life of a subject which has been treated for cancer. In one embodiment, survival refers to the failure of a tumor to recur. As used herein, the term "recur" refers to the re-growth of tumor or cancerous cells in a subject in whom primary treatment for the tumor has been administered. The tumor may recur in the original site or in another part of the body. In one embodiment, a tumor that recurs is of the same type as the original tumor for which the subject was treated. For example, if a subject had an ovarian cancer tumor, was treated and subsequently developed another ovarian cancer tumor, the tumor has recurred. In addition, a cancer can recur in a different organ or tissue than the one where it originally occurred.
  • II. Methods and Kits of the Invention
  • The present invention is based, at least in part, on the unexpected discovery that folate receptor alpha (FRα), not bound to a cell, is found at elevated levels in the body fluids, for example, urine or serum, of a subject having an FRα-expressing cancer as compared to a control sample. Moreover, the present invention is based, at least in part, on the identification of an immunological assay that exhibits the necessary sensitivity for assessing levels of FRα not bound to a cells in samples, where prior attempts to do so had repeatedly failed. Indeed, the present invention overcomes the challenges observed during prior attempts to develop an FRα-based diagnostic assay for FRα-expressing cancer such as lung or ovarian cancer by providing an immunological assay capable of accurately assessing levels of FRα not bound to a cell in a sample, e.g., urine or serum.
  • Accordingly, methods and kits for assessing whether a subject has or is at risk for developing an FRα-expressing cancer and, further, for assessing the progression of an FRα-expressing cancer are provided. In various embodiments, the methods involve the comparison of levels of FRα not bound to a cell in samples, for example, urine and serum, as compared to control levels, in assessing the presence, degree or risk of development of ovarian cancer in the subject.
  • A. Diagnostic Method, Prognostic Methods, Risk Assessment Methods, and Stratification Methods,
  • Specifically, the present invention provides diagnostic methods for assessing whether a subject is afflicted with an FRα-expressing cancer, such as lung or ovarian cancer, prognostic methods for predicting the progression of an FRα-expressing cancer such as lung or ovarian cancer, and risk assessment methods for assessing the level of risk that a subject will develop the FRα-expressing cancer. Furthermore, the invention provides stratification methods for stratifying an FRα-expressing cancer such as lung or ovarian cancer subject into cancer therapy groups. The various aspects and embodiments of the invention discussed here are intended to be non-limiting and to encompass all possible combinations of the specific embodiments mentioned, which may apply to any of the methods and kits discussed herein or claimed below.
  • The methods of the present invention can be practiced in conjunction with any other method used by the skilled practitioner to diagnose an FRα-expressing cancer, predict the progression of an FRα-expressing cancer, or to assess the level of risk that a subject will develop an FRα-expressing cancer.
  • In one aspect, the invention provides a method of assessing whether a subject is afflicted with an FRα-expressing cancer, by determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample (such as urine or serum) derived from the subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein a difference between the level of FRα in the sample derived from the subject and the level of FRα in the control sample is an indication that the subject is afflicted with an FRα-expressing cancer. In a particular embodiment, the level of FRα in the sample derived from the subject is assessed by contacting the sample with an antibody that binds FRα not bound to a cell and is selected from the group consisting of (a) an antibody that binds the same epitope as the MORAb-003 antibody; and (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. In one embodiment, the sample is selected from the group consisting of urine and serum.
  • In another aspect, the present invention provides a method of assessing whether a subject is afflicted with an FRα-expressing cancer such as lung cancer or ovarian cancer, the method comprising determining the level of folate receptor alpha (FRα) not bound to a cell in a urine sample derived from the subject; and comparing the level of folate receptor alpha (FRα) in the urine sample derived from the subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the urine sample derived from the subject and the level of FRα in the control sample is an indication that the subject is afflicted with an FRα-expressing cancer.
  • In another aspect, the invention provides a method of assessing whether a subject is afflicted with an FRα-expressing cancer, by determining the level of folate receptor alpha (FRα) not bound to a cell in a serum sample derived from the subject; and comparing the level of folate receptor alpha (FRα) in the serum sample derived from the subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the serum sample derived from the subject and the level of FRα in the control sample is an indication that the subject is afflicted with an FRα-expressing cancer. In particular embodiments, the subject has not been treated with an agent, such as a steroid, that enhances the levels of FRα in serum. In a specific embodiment, the FRα-expressing cancer is ovarian cancer and the subject has not been treated with an agent, such as a steroid, that enhances the levels of FRα in serum.
  • In the methods and kits of the present invention, FRα-expressing cancers include cancers characterized in that the cancer cells express FRa. In particular embodiments, the FRα is released from the cancer cells, for example, from the surface of the cancer cell, and into the biological fluids of the subject. FRα-expressing cancers include lung cancer (e.g., bronchioalveolar carcinomas, carcinoid tumors, and non-small cell lung cancers, such as adenocarcinomas); mesothelioma; ovarian cancer; renal cancer; brain cancer (e.g., anaplastic ependymoma and cerebellar juvenile pilocytic astrocytoma); cervical cancer; nasopharyngeal cancer; mesodermally derived tumor; squamous cell carcinoma of the head and neck; endometrial cancer; endometrioid adenocarcinomas of the ovary, serous cystadenocarcinomas, breast cancer; bladder cancer; pancreatic cancer; bone cancer (e.g., high-grade osteosarcoma); and pituitary cancer (e.g., pituitary adenoma). In a particular embodiment, the FRα-expressing cancer is ovarian cancer.
  • In certain embodiments of the methods and kits of the present invention, the FRα-expressing cancer is lung cancer. In more specific embodiments, the lung cancer is non-small cell lung carcinoma (NSCLC). In one such embodiment, the NSCLC is selected from the group consisting of adenocarcinoma, squamous cell lung carcinoma, large cell lung carcinoma, pleomorphic NSCLC, carcinoid tumor, salivary gland carcinoma, and unclassified carcinoma. In a preferred embodiment, the NSCLC is adenocarcinoma. In alternative embodiments, the lung cancer is small cell lung carcinoma (SCLC). In another embodiment, the lung cancer is bronchioalveolar carcinoma. In yet another embodiment, the lung cancer is a lung carcinoid tumor.
  • The present invention also provides methods to assess whether a subject is afflicted with ovarian cancer by determining the level of folate receptor alpha (FRα) not bound to a cell in a urine sample derived from the subject, wherein the presence of FRα in the urine sample at a concentration of greater than about 3000 a.u./ml is an indication that the subject is afflicted with ovarian cancer. In particular embodiments, the presence of FRα in the urine sample at a concentration of greater than about 4000 a.u./ml, about 5000 a.u./ml, about 6000 a.u./ml, about 7000 a.u./ml, about 8000 a.u./ml, about 9000 a.u./ml, about 10,000 a.u./ml, about 11,000 a.u./ml, about 12,000 a.u./ml, about 13,000 a.u./ml, about 14,000 a.u./ml, about 15,000 a.u./ml, about 16,000 a.u./ml, about 17,000 a.u./ml, about 18,000 a.u./ml, about 19,000 a.u./ml, about 20,000 a.u./ml, about 21,000 a.u./ml, about 22,000 a.u./ml, about 23,000 a.u./ml, about 24,000 a.u./ml, about 25,000 a.u./ml, about 26,000 a.u./ml, about 27,000 a.u./ml, about 28,000 a.u./ml, about 29,000 a.u./ml or about 30,000 a.u./ml is an indication that the subject is afflicted with ovarian cancer.
  • In yet another aspect, the present invention provides a method of assessing whether a subject is afflicted with ovarian cancer, by determining the level of folate receptor alpha (FRα) in a urine sample derived from the subject, wherein the presence of FRα in the urine sample at a concentration of greater than about 9100 pg/ml is an indication that the subject is afflicted with ovarian cancer or wherein a concentration of less than about 9100 pg/ml is an indication that the subject is not afflicted with ovarian cancer. For example, the presence of FRα in the urine sample at a concentration of greater than about 9500 pg/mL, about 10,000 pg/mL, about 11,000 pg/mL, about 12,000 pg/mL, about 13,000 pg/mL, about 14,000 pg/mL, about 15,000 pg/mL, about 16,000 pg/mL, about 17,000 pg/mL, about 18,000 pg/mL, about 19,000 pg/mL, about 20,000 pg/mL, about 21,000 pg/mL, about 22,000 pg/mL, about 23,000 pg/mL, about 24,000 pg/mL, about 25,000 pg/mL, about 26,000 pg/mL, about 27,000 pg/mL, about 28,000 pg/mL, about 29,000 pg/mL, about 30,000 pg/mL, about 40,000 pg/mL, about 50,000 pg/mL, about 60,000 pg/mL, about 70,000 pg/mL, about 80,000 pg/ml, about 90,000 pg/ml, about 100,000 pg/ml or about 150,000 pg/ml is an indication that the subject is afflicted with ovarian cancer.
  • In certain embodiments of the foregoing aspects of the invention, the levels of FRα not bound to a cell in a sample (e.g., a sample such as a urine sample or serum sample) derived from a subject are compared with the levels of FRα in a control sample, wherein a difference between the levels is an indication that the subject is afflicted with an FRα-expressing cancer such as lung or ovarian cancer. In a particular embodiment, the difference constitutes an increase in the level of FRα not bound to a cell in the sample derived from the subject as compared with the level of FRα in the control sample, wherein this increase is indicative of the presence or growth of FRα-expressing cancer. Alternatively, the difference constitutes a decrease in the level of FRα, wherein the decrease is indicative of the absence or regression of FRa-expressing cancer. As used herein, "a difference" between the level of folate receptor alpha not bound to a cell in a sample from a subject (i.e., a test sample) and the level of folate receptor alpha in a control sample refers broadly to any clinically relevant change (including an increase or a decrease) and/or statistically significant difference in the level of folate receptor alpha in the two samples. In an exemplary embodiment, the difference is selected based on a cutoff value determined using a receiver operating characteristic (ROC) analysis, an example of which is presented in Example 6. The optimal cutoff value may vary depending on the assay methods and conditions employed. In other embodiments, the difference must be greater than the limits of detection of the method for determining the level of FRα not bound to a cell. It is preferred that the difference be at least greater than the standard error of the assessment method, and preferably a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method. The difference may be assessed by any appropriate comparison, including any appropriate parametric or nonparametric descriptive statistic or comparison. For example, "an increase" in the level of FRα may refer to a level that exceeds a cutoff value determined using an ROC analysis. It may also refer to a level in a test sample that is two, and more preferably about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900% or about 1000% more than the level of FRα in the control sample. An increase may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations above the average level of FRα in the control sample. Likewise, "a decrease" in the level of FRα not bound to a cell may refer to a level in a test sample that does not exceed a cutoff value determined using an ROC analysis. It may also refer to a level in a test sample that is about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, or about 90% less than the level of FRα in the control sample. A decrease may also refer to a level in a test sample that is preferably at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations below the average level of FRα in the control sample.
  • Samples useful in the methods and kits of the invention include any tissue, cell, biopsy, or bodily fluid that may contain detectable levels of FRα not bound to a cell. In one embodiment, a sample may be a tissue, a cell, whole blood, plasma, buccal scrape, saliva, cerebrospinal fluid, stool, or bronchoalveolar lavage. In some embodiments, the sample is FRα-expressing tumor sample or a sample of tissues or cells where FRα-expressing cancer may be found. In preferred embodiments, the sample is a urine or serum sample.
  • Body samples may be obtained from a subject by a variety of techniques known in the art including, for example, by the use of a biopsy or by scraping or swabbing an area or by using a needle to aspirate bodily fluids. Methods for collecting various body samples are well known in the art.
  • Samples suitable for detecting and quantitating the FRα protein level may be fresh, frozen, or fixed according to methods known to one of skill in the art. Suitable tissue samples are preferably sectioned and placed on a microscope slide for further analyses. Solid samples, i.e., tissue samples, may be solubilized and/or homogenized and subsequently analyzed as soluble extracts. Liquid samples may also be subjected to physical or chemical treatments. In some embodiments, urine samples are treated by centrifugation, vortexing, dilution and/or treatment with a solubilizing substance (such as, for example, guanidine treatment).
  • In one embodiment, a freshly obtained biopsy sample is frozen using, for example, liquid nitrogen or difluorodichloromethane. The frozen sample is mounted for sectioning using, for example, OCT, and serially sectioned in a cryostat. The serial sections are collected on a glass microscope slide. For immunohistochemical staining the slides may be coated with, for example, chrome-alum, gelatine or poly-L-lysine to ensure that the sections stick to the slides. In another embodiment, samples are fixed and embedded prior to sectioning. For example, a tissue sample may be fixed in, for example, formalin, serially dehydrated and embedded in, for example, paraffin.
  • Once the sample is obtained, any method known in the art to be suitable for detecting and quantitating FRα not bound to a cell may be used (either at the nucleic acid or, preferably, at the protein level), as described in section (B) below. Exemplary methods are well known in the art and include but are not limited to western blots, northern blots, southern blots, immunohistochemistry, solution phase assay, ELISA, e.g., amplified ELISA, immunoprecipitation, immunofluorescence, flow cytometry, immunocytochemistry, mass spectrometrometric analyses, e.g., MALDI-TOF and SELDI-TOF, nucleic acid hybridization techniques, nucleic acid reverse transcription methods, and nucleic acid amplification methods.
  • In many embodiments, the level of FRα not bound to a cell in the sample (such as, for example, urine or serum) is assessed by contacting the sample with an antibody that binds FRa. Antibodies that bind FRα are known in the art and include (i) the murine monoclonal LK26 antibody (the heavy and light chains thereof are presented herein as SEQ ID NOs: 22 and 23), as described in European Patent Application No. 86104170.5 (Rettig) (the entire contents of which are incorporated herein by reference); (ii) the MORAB-003 antibody, as described in International Publication No. WO2004/113388 and U.S. Patent No. 5,646,253 , the entire contents of each of which are incorporated herein by reference. The monoclonal antibodies MOV18 and MOv19 also bind different epitopes on the FRα molecule (previously known as gp38/FBP). Miotti, S. et al. Int J Cancer, 38: 297-303 (1987). For example, the MOV18 antibody binds the epitope set forth herein as SEQ ID NO:26 (TELLNVXMNAK*XKEKPXPX*KLXXQX) (note that at position 12, a tryptophan or histidine residue is possible, and at position 21, an aspartic acid or glutamic acid residue is possible), as taught in Coney et al. Cancer Res, 51: 6125-6132 (1991).
  • As used herein, the term "MORAb-003" refers to an antibody that specifically binds FRα and which comprises the mature heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the mature light chain sequence of SEQ ID NO:8. The corresponding pre-protein amino acid sequences for MORAb-003 are set forth in SEQ ID NOs: 9 (heavy chain) and 10 (light chain). The MORAb-003 antibody comprises the following CDRs: SEQ ID NO:1 as CDRH1, SEQ ID NO:2 as CDRH2, SEQ ID NO:3 as CDRH3, SEQ ID NO:4 as CDRL1, SEQ ID NO:5 as CDRL2, and SEQ ID NO:6 as CDRL3. MORAb-003 antibody producing cells have been deposited with the American Type Culture Collection (10801 University Blvd., Manassas, Virginia 20110-2209) on April 24, 2006 and have been assigned Accession No. PTA-7552.
  • Other antibodies that bind FRα and for use in the methods of the present invention include 9F3.H9.H3.H3.B5.G2 (also referred to as 9F3),19D4.B7 (also referred to as 19D4), 24F12.B1 (also referred to as 24F12), and 26B3.F2 (also referred to as 26B3). The amino acid sequences of these antibodies, their CDRs, and their heavy and light chain variable domains, as well as polynucleotide sequences that may encode them, are provided in Table 33. In some embodiments, these antibodies are murine IgG, or derivatives thereof. In other embodiments, the antibodies are human, humanized, or chimeric.
  • 9F3
  • In some embodiments, the antibody that binds FRα is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:27. In some embodiments, the antibody that binds FRα includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:28. In some embodiments, the antibody that binds FRα includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO 29. In some embodiments, the antibody that binds FRα includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO 31. In some embodiments, the antibody that binds FRα includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:32. In some embodiments, the antibody that binds FRα includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:33. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:27; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:28; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:29. The antibody that binds FRα may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:31; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO: 32; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:33. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:27; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:28; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:29, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:31; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:32; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:33.
  • The antibody that binds FRα may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:30. The antibody that binds FRα may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:34. The antibody that binds FRα may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:30, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:34. In some embodiments the antibody that binds FRα is the 9F3.H9.H3.H3.B5.G2 (9F3) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FRα. In some embodiments, the antibody has a murine IgG2a constant region.
  • In some embodiments, the antibody that binds FRα is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Blvd., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11887. In some embodiments, the antibody that binds FRα comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells. In some embodiments, antibody that binds FRα comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • 19D4
  • In some embodiments, the antibody that binds FRα is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:35. In some embodiments, the antibody that binds FRα includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:36. In some embodiments, the antibody that binds FRα includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:37. In some embodiments, the antibody that binds FRα includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:39. In some embodiments, the antibody that binds FRα includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:40. In some embodiments, the antibody that binds FRα includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:41. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:35; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:36; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:37. The antibody that binds FRα may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:39; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO: 40; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:41. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:35; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:36; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:37, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:39; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:40; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:41.
  • The antibody that binds FRα may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:38. The antibody that binds FRα may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:42. The antibody that binds FRα may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:38, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:42. In some embodiments, the antibody that binds FRα is the 19D4.B7 (19D4) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FRα. In some embodiments, the antibody has a murine IgG2a constant region.
  • In some embodiments, the antibody that binds FRα is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Blvd., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11884. In some embodiments, the antibody that binds FRα comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells. In some embodiments, antibody that binds FRα comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • 24F12
  • In some embodiments, the antibody that binds FRα is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:43. In some embodiments, the antibody that binds FRα includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:44. In some embodiments, the antibody that binds FRα includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:45. In some embodiments, the antibody that binds FRα includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:47. In some embodiments, the antibody that binds FRα includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:48. In some embodiments, the antibody that binds FRα includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:49. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:43; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:44; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:45. The antibody that binds FRα may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:47; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO: 48; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:49. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:43; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:44; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:45, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:47; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:48; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:49.
  • The antibody that binds FRα may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:46. The antibody that binds FRα may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:50. The antibody that binds FRα may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:46, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:50. In some embodiments the antibody that binds FRα is the 24F12.B1 (24F12) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FRa. In some embodiments, the antibody has a murine IgG1 constant region.
  • In some embodiments, the antibody that binds FRα is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Blvd., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11886. In some embodiments, the antibody that binds FRα comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells. In some embodiments, antibody that binds FRα comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • 26B3
  • In some embodiments, the antibody that binds FRα is an antibody or antigen-binding fragment that includes a light chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:51. In some embodiments, the antibody that binds FRα includes a light chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:52. In some embodiments, the antibody that binds FRα includes a light chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:53. In some embodiments, the antibody that binds FRα includes a heavy chain CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:55. In some embodiments, the antibody that binds FRα includes a heavy chain CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:56. In some embodiments, the antibody that binds FRα includes a heavy chain CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:57. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:51; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:52; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:53. The antibody that binds FRα may include a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:55; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:56; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:57. The antibody that binds FRα may include a light chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:51; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:52; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:53, and also have a heavy chain having a CDR1 amino acid sequence substantially the same as, or identical to, SEQ ID NO:55; a CDR2 amino acid sequence substantially the same as, or identical to, SEQ ID NO:56; and a CDR3 amino acid sequence substantially the same as, or identical to, SEQ ID NO:57.
  • The antibody that binds FRα may include a light chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:54. The antibody that binds FRα may include a heavy chain variable domain that includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:58. The antibody that binds FRα may include a light and a heavy chain variable domains, wherein the light chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:54, and the heavy chain variable domain includes an amino acid sequence substantially the same as, or identical to, SEQ ID NO:58. In some embodiments the antibody that binds FRα is the 26B3.F2 (26B3) antibody or an antigen-binding fragment thereof, capable of binding either a native or nonreduced form of FRα. In some embodiments, the antibody has a murine IgG1 constant region.
  • In some embodiments, the antibody that binds FRα is an antibody that is produced by antibody-producing cells deposited with the American Type Culture Collection (10801 University Blvd., Manassas, Virginia 20110-2209) on May 19, 2011 and have been assigned Accession No. PTA-11885. In some embodiments, the antibody that binds FRα comprises one or more of the light and heavy chain CDRs of the antibodies produced by the deposited antibody-producing cells. In some embodiments, antibody that binds FRα comprises the light and heavy chain variable regions of the antibodies produced by the deposited antibody-producing cells.
  • Antigen binding arrangements of CDRs may be engineered using antibody-like proteins as CDR scaffolding. Engineered antigen-binding proteins are included within the scope of antibodies that bind FRa.
  • Other reagent antibodies that bind FRα are known in the art, and presently, multiple such reagent antibodies are commercially available (based on search of anti-FRα antibodies at http://www.biocompare.com), as listed in the table below.
    Product Company Quantity Applications Reactivity
    Mouse Anti-Human FRα Purified - MaxPab Polyclonal Antibody, Unconjugated Abnova Corporation 50 µg Detection Antibody, Western Blot (Transfected lysate) Human
    Mouse Anti-Human FRα Purified MaxPab Polyclonal Antibody, Unconjugated Abnova Corporation 50 µg Detection Antibody, Western Blot (Transfected lysate) Human
    Rabbit Anti-Human FRα Purified MaxPab Polyclonal Antibody, Abnova Corporation 100 µg Detection Antibody, Western Blot (Transfected lysate) Human
    Rabbit Anti-FRa Polyclonal Antibody, Unconjugated Aviva Systems Biology 50 µg Western Blot Human, Mouse, Rat
    Rabbit Anti-Human FRα Polyclonal Antibody, Unconjugated GeneTex 100 µL Western blot. The usefulness of this product in other applications has not been determined. Human
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Biotin Conjugated LifeSpan BioSciences 10 mg ELISA (1:4000 - 1:20000), Immunofluorescence, Immunohistochemistry, Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Biotin Conjugated LifeSpan BioSciences Not provided ELISA (1:5000 - 1:25000), Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Hrp Conjugated LifeSpan BioSciences 20 mg ELISA (1:2000 - 1:10000), Immunohistochemistry, Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Hrp Conjugated LifeSpan BioSciences 1000 µg ELISA (1:2000 - 1:12000), Gel Shift, Immunohistochemistry (1:100 - 1:200), Immunohistochemistry Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Hrp Conjugated LifeSpan BioSciences 2000 µg (200 µl) ELISA, Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Hrp Conjugated, LifeSpan BioSciences Not provided ELISA, Immunohistochemistry (Frozen sections), Immunohistochemistry (Parrafin), Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Unconjugated LifeSpan BioSciences 50 mg ELISA (1:10000 - 1:40000), Immunoprecipitation, Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Unconjugated LifeSpan BioSciences 10000 µg ELISA (1:10000 - 1:40000), Immunoprecipitation, Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Unconjugated LifeSpan BioSciences 1 ml ELISA (1:3000 - 1:9000), Immunoprecipitation, Western Blot Bovine
    Goat Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Unconjugated Folate Receptor Alpha (FRα) LifeSpan BioSciences Not provided ELISA (1:3000 - 1:9000), Immunoprecipitation, Western Blot Bovine
    Mouse Anti-Bovine Folate Receptor Alpha (FRα) Monoclonal, Unconjugated LifeSpan BioSciences 200 µg ELISA Bovine
    Mouse Anti-Bovine Folate Receptor Alpha LifeSpan BioSciences 200 µg ELISA Human
    (FRα) Monoclonal, Unconjugated Folate Receptor Alpha (FRα)
    Mouse Anti-Bovine Folate Receptor Alpha (FRα) Monoclonal, Unconjugated LifeSpan BioSciences 200 µg ELISA Human
    Mouse Anti-Bovine Folate Receptor Alpha (FRα) Monoclonal, Unconjugated LifeSpan BioSciences 200 µg ELISA Not provided
    Mouse Anti-Human Folate Receptor Alpha (FRα) Monoclonal, Unconjugated, Clone 6d398 LifeSpan BioSciences 100 µl ELISA (1 - 10 µg/ml), Flow Cytometry, Immunocytochemistry, Immunohistochemistry (Frozen sections) Monkey
    Rabbit Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Unconjugated LifeSpan BioSciences 1 ml ELISA Bovine
    Rabbit Anti-Bovine Folate Receptor Alpha (FRα) Polyclonal, Unconjugated LifeSpan BioSciences Not provided Not provided Bovine
    Mouse Anti-Human FRα Polyclonal antibody, Unconjugated, Clone folate receptor 1 (adult) Novus Biologicals 0.05 ml Western Blot, ELISA
    Mouse Anti-Human FRα Polyclonal, Unconjugated Novus Biologicals 0.05 mg ELISA, Western Blot Human
    Goat Anti-Human FRα Affinity purified Polyclonal antibody, Biotin Conjugated R&D Systems 50 µg Western Blot Human
    Goat Anti-Human FRα Affinity purified Polyclonal antibody, Unconjugated R&D Systems 100 µg Flow Cytometry, Western Blot Human
    Mouse Anti-Human FRα Monoclonal Antibody, Allophycocyanin Conjugated, Clone 548908 R&D Systems 100 Tests Flow Cytometry Human
    Mouse Anti-Human FRα Monoclonal Antibody, Phycoerythrin Conjugated, Clone 548908 R&D Systems 100 Tests Flow Cytometry Human
    Mouse Anti-Human FRα Monoclonal antibody, Unconjugated, Clone 548908 R&D Systems 100 µg Flow Cytometry, Immunocytochemistry, Western Blot Human
    Mouse Anti- Human FRα Monoclonal Antibody, Unconjugated United States Biological 100 µg ELISA, Flow Cytometry, Immunocytochemistry, Western Blot Human
  • In a preferred embodiment, the antibody that binds FRα comprises at least one of the following CDRs, as derived from the murine LK26 heavy and light chains: SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3. See US Patent No. 5,646,253 , the contents of which, as they relate to the anti-FRα antibodies that may be used in the present invention, are incorporated herein by reference. Further mutations may be made in the framework regions as taught in US Patent No. 5,646,253 , the contents of which are hereby incorporated by reference.
  • In another preferred embodiment, the antibody includes a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13) LK26HuVKY (SEQ ID NO: 14), LK26HuVKPW (SEQ ID NO: 15), and LK26HuVKPW,Y (SEQ ID NO: 16); and a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21). See US Patent No. 5,646,253 and US Patent No. 6,124,106 . In another embodiment, the antibody comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16). In another embodiment, the antibody comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16). In a further embodiment, the antibody comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  • In some embodiments, samples may need to be modified in order to make FRα accessible to antibody binding. In a particular aspect of the immunocytochemistry or immunohistochemistry methods, slides may be transferred to a pretreatment buffer and optionally heated to increase antigen accessibility. Heating of the sample in the pretreatment buffer rapidly disrupts the lipid bi-layer of the cells and makes the antigens (may be the case in fresh specimens, but not typically what occurs in fixed specimens) (i.e., the FRα protein) more accessible for antibody binding. The term "pretreatment buffer" are used interchangeably herein to refer to a buffer that is used to prepare cytology or histology samples for immunostaining, particularly by increasing FRα protein accessibility for antibody binding. The pretreatment buffer may comprise a pH-specific salt solution, a polymer, a detergent, or a nonionic or anionic surfactant such as, for example, an ethyloxylated anionic or nonionic surfactant, an alkanoate or an alkoxylate or even blends of these surfactants or even the use of a bile salt. The pretreatment buffer may, for example, be a solution of 0.1 % to 1% of deoxycholic acid, sodium salt, or a solution of sodium laureth-13-carboxylate (e.g., Sandopan LS) or and ethoxylated anionic complex. In some embodiments, the pretreatment buffer may also be used as a slide storage buffer. In a particular embodiment, the sample, for example, the urine sample, is centrifuged, vortexed, diluted and/or subjected to guanidine treatment.
  • Any method for making FRα protein more accessible for antibody binding may be used in the practice of the invention, including the antigen retrieval methods known in the art. See, for example, Bibbo, et al. (2002) Acta. Cytol. 46:25-29; Saqi, et al. (2003) Diagn. Cytopathol. 27:365-370; Bibbo, et al. (2003) Anal. Quant. Cytol. Histol. 25:8-11, the entire contents of each of which are incorporated herein by reference.
  • Following pretreatment to increase FRα protein accessibility, samples may be blocked using an appropriate blocking agent, e.g., a peroxidase blocking reagent such as hydrogen peroxide. In some embodiments, the samples may be blocked using a protein blocking reagent to prevent non-specific binding of the antibody. The protein blocking reagent may comprise, for example, purified casein. An antibody, particularly a monoclonal or polyclonal antibody, that specifically binds to FRα is then incubated with the sample.
  • Techniques for detecting antibody binding are well known in the art. Antibody binding to FRα may be detected through the use of chemical reagents that generate a detectable signal that corresponds to the level of antibody binding and, accordingly, to the level of FRα protein expression. In one of the immunohistochemistry or immunocytochemistry methods of the invention, antibody binding is detected through the use of a secondary antibody that is conjugated to a labeled polymer. Examples of labeled polymers include but are not limited to polymer-enzyme conjugates. The enzymes in these complexes are typically used to catalyze the deposition of a chromagen at the antigen-antibody binding site, thereby resulting in cell staining that corresponds to expression level of the biomarker of interest. Enzymes include, but are not limited to, horseradish peroxidase (HRP) and alkaline phosphatase (AP).
  • In one immunohistochemistry or immunocytochemistry method of the invention, antibody binding to the FRα protein is detected through the use of an HRP-labeled polymer that is conjugated to a secondary antibody. Antibody binding can also be detected through the use of a species-specific probe reagent, which binds to monoclonal or polyclonal antibodies, and a polymer conjugated to HRP, which binds to the species specific probe reagent. Slides are stained for antibody binding using any chromagen, e.g., the chromagen 3,3-diaminobenzidine (DAB), and then counterstained with hematoxylin and, optionally, a bluing agent such as ammonium hydroxide or TBS/Tween-20. Other suitable chromagens include, for example, 3-amino-9-ethylcarbazole (AEC). In some aspects of the invention, slides are reviewed microscopically by a cytotechnologist and/or a pathologist to assess cell staining, e.g., fluorescent staining (i.e., FRα expression). Alternatively, samples may be reviewed via automated microscopy or by personnel with the assistance of computer software that facilitates the identification of positive staining cells.
  • In a preferred embodiment of the invention, the antibody is labeled. For example, detection of antibody binding can be facilitated by coupling the anti-FRα antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include 125I, 131T, 35S, 14C, or 3H. In a particular embodiment, the antibody is labeled with a radio-label, chromophore-label, fluorophore-label, or enzyme-label.
  • In one embodiment of the invention frozen samples are prepared as described above and subsequently stained with antibodies against FRα diluted to an appropriate concentration using, for example, Tris-buffered saline (TBS). Primary antibodies can be detected by incubating the slides in biotinylated anti-immunoglobulin. This signal can optionally be amplified and visualized using diaminobenzidine precipitation of the antigen. Furthermore, slides can be optionally counterstained with, for example, hematoxylin, to visualize the cells.
  • In another embodiment, fixed and embedded samples are stained with antibodies against FRα and counterstained as described above for frozen sections. In addition, samples may be optionally treated with agents to amplify the signal in order to visualize antibody staining. For example, a peroxidase-catalyzed deposition of biotinyl-tyramide, which in turn is reacted with peroxidase-conjugated streptavidin (Catalyzed Signal Amplification (CSA) System, DAKO, Carpinteria, CA) may be used.
  • One of skill in the art will recognize that the concentration of a particular antibody used to practice the methods of the invention will vary depending on such factors as time for binding, level of specificity of the antibody for FRα, and method of sample preparation. Moreover, when multiple antibodies are used, the required concentration may be affected by the order in which the antibodies are applied to the sample, e.g., simultaneously as a cocktail or sequentially as individual antibody reagents. Furthermore, the detection chemistry used to visualize antibody binding to FRα must be optimized to produce the desired signal to noise ratio.
    In one embodiment of the invention, proteomic methods, e.g., mass spectrometry, are used for detecting and quantitating the FRα protein. For example, matrix-associated laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) or surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF MS) which involves the application of a sample, such as serum, to a protein-binding chip (Wright, G.L., Jr., et al. (2002) Expert Rev Mol Diagn 2:549; Li, J., et al. (2002) Clin Chem 48:1296; Laronga, C., et al. (2003) Dis Markers 19:229; Petricoin, E.F., et al. (2002) 359:572; Adam, B.L., et al. (2002) Cancer Res 62:3609; Tolson, J., et al. (2004) Lab Invest 84:845; Xiao, Z., et al. (2001) Cancer Res 61:6029) can be used to detect and quantitate the FRα protein. Mass spectrometric methods are described in, for example, U.S. Patent Nos. 5,622,824 , 5,605,798 and 5,547,835 , the entire contents of each of which are incorporated herein by reference.
  • The present invention is further predicated, at least in part, on the identification of FRα as a prognostic biomarker, i.e., as a biomarker of the progression and/ or severity, of an FRα-expressing cancer such as ovarian cancer or non-small cell lung cancer. Accordingly, the present invention provides methods of assessing the progression of an FRα-expressing cancer in a subject afflicted with ovarian cancer by comparing the level of FRα in a sample derived from a subject with the level of FRα in a control sample, wherein a difference in the level of FRα in the sample (such as a urine or serum sample) derived from the subject compared with the control sample is an indication that the cancer will progress rapidly. Similarly, methods of assessing the level of risk that a subject will develop an FRα-expressing cancer involve comparing the level of FRα in a sample derived from a subject with the level of FRα in a control sample, wherein a difference in the level of FRα in the sample (such as urine or serum sample) derived from the subject compared with the control sample is an indication that the subject has a higher level of risk of developing an FRα-expressing cancer as compared to normal risk in a healthy individual.
  • In one embodiment, the difference is an increase. In another embodiment, the difference is a decrease. In some types of cancers (e.g., squamous cell carcinoma of the head and neck, ovarian cancer), a higher level of FRα expression is associated with a worse prognosis, whereas in other types of cancers (e.g., non-small-cell lung cancers), a higher level of FRα expression is associated with a better prognosis. Thus, in one specific embodiment, the FRα-expressing cancer is ovarian cancer or squamous cell carcinoma of the head and neck and the difference is an increase. In another specific embodiment, the FRα-expressing cancer is a non small-cell lung cancer, and the difference is a decrease.
  • In certain aspects, the invention provides methods of assessing the progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer by comparing the level of FRα in a sample derived from a subject with the level of FRα in a control sample, wherein an increase in the level of FRα in the sample (such as a urine or serum sample) derived from the subject compared with the control sample is an indication that the cancer will progress rapidly, or a decrease in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the cancer will progress slowly or will regress. Similarly, methods of assessing the level of risk that a subject will develop an FRα-expressing cancer involve comparing the level of FRα in a sample derived from a subject with the level of FRα in a control sample, wherein an increase in the level of FRα in the sample (such as urine or serum sample) derived from the subject compared with the control sample is an indication that the subject has a higher level of risk of developing an FRα-expressing cancer as compared to normal risk in a healthy individual, or a decrease in the level of FRα in the sample derived from the subject as compared with the level of FRα in the control sample is an indication that the subject has a lower level of risk of developing an FRα-expressing cancer as compared to a normal risk in a healthy individual.
  • Any clinically relevant or statistically significant increase or decrease, using any analytical method known in the art, may be utilized in the prognostic, risk assessment and other methods of the invention. In one embodiment, an increase in the level of FRα in the level of FRα refers to a level that exceeds a cutoff value determined using an ROC analysis as exemplified in Example 6. In another embodiment, a decrease in the level of FRα refers to a level in a test sample that does not exceed a cutoff value determined using an ROC analysis.
  • In other embodiments, the increase or decrease must be greater than the limits of detection of the method for determining the level of FRα. In further embodiments, the increase or decrease be at least greater than the standard error of the assessment method, and preferably a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method. In some embodiments, the increase or decrease is assessed using parametric or nonparametric descriptive statistics, comparisons, regression analyses, and the like.
  • In other embodiments, the increase or decrease is a level in the sample derived from the subject that is about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900% or about 1000% more or less than the level of FRα in the control sample. In alternative embodiments, the increase or decrease is a level in the sample derived from the subject that is at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations above or below the average level of FRα in the control sample. As used herein, the phrase "progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer" may refer to the progression of an FRα-expressing cancer from a less severe to a more severe cancer state. This could include an increase in the number or severity of tumors, the degree of metastasis, the speed with which the cancer is growing and spreading, and the like. In certain embodiments, the progression is a progression from a less severe stage to a more severe stage, wherein the stage is assessed according to a staging scheme known in the art. In one embodiment, wherein the FRα-expressing cancer is ovarian cancer, the progression refers to a progression from Stage I to Stage II, from Stage II to Stage III, etc. In another embodiment, wherein the FRα-expressing cancer is non-small cell lung cancer (NSCLC), the progression refers to a progression from Stage 0 to Stage IA, Stage IA to Stage IB, Stage IB to Stage IIA, Stage IIA to Stage IIB, Stage IIB to Stage IIC, etc. In another embodiment, wherein the FRα-expressing cancer is non-small cell lung cancer (NSCLC), the progression refers to a progression from a less severe to a more severe stage as determined under the TNM classification system. See Spira; Greene; Sobin.
  • Alternatively, the phrase "progression of an FRα-expressing cancer in a subject afflicted with an FRα-expressing cancer" may refer to a regression of an FRα-expressing cancer from a more severe state to a less severe state, such as a decrease in the number or severity of tumors, the degree of metastasis, the speed with which the cancer is growing and spreading, and the like. In certain embodiments, the progression is a progression from a more severe stage to a less severe stage, wherein the stage is assessed according to a staging scheme known in the art. In one embodiment, wherein the FRα-expressing cancer is ovarian cancer, the progression refers to a regression from Stage IV to Stage III, from Stage III to Stage II, etc. In another embodiment, wherein the FRα-expressing cancer is non-small cell lung cancer (NSCLC), the progression refers to a progression from Stage IV to Stage IIIB, Stage IIIB to Stage IIIA, Stage IIIA to Stage IIB, etc. In another embodiment, wherein the FRα-expressing cancer is non-small cell lung cancer (NSCLC), the progression refers to a progression from a more severe to a less severe stage as determined under the TNM classification system. See Spira; Greene; Sobin.
  • In further embodiments, the level of FRα may be used to calculate the likelihood that a subject is afflicted with an FRα-expressing cancer, the progression of an FRα-expressing cancer in a subject, the level of risk of developing an FRα-expressing cancer, the risk of cancer recurrence in a subject being treated for an FRα-expressing, the survival of a subject being treated for an FRα-expressing cancer, the efficacy of a treatment regimen for treating an FRα-expressing cancer, and the like, using the methods of the invention, which may include methods of regression analysis known to one of skill in the art. For example, suitable regression models include, but are not limited to CART (e.g., Hill, T, and Lewicki, P. (2006) "STATISTICS Methods and Applications" StatSoft, Tulsa, OK), Cox (e.g., www.evidence-based-medicine.co.uk), exponential, normal and log normal (e.g., www.obgyn.cam.ac.uk/mrg/statsbook/stsurvan.html), logistic (e.g., www.en.wikipedia.org/wiki/Logistic_regression), parametric, non-parametric, semi-parametric (e.g., www.socserv.mcmaster.ca/jfox/Books/Companion), linear (e.g., www.en.wikipedia.org/wiki/Linear_regression), or additive (e.g., www.en.wikipedia.org/wiki/Generalized_additive_model).
  • In one embodiment, a regression analysis includes the level of FRα. In further embodiments, a regression analysis may include additional clinical and/or molecular co-variates. Such clinical co-variates include, but are not limited to, age of the subject, tumor stage, tumor grade, tumor size, treatment regime, e.g., chemotherapy and/or radiation therapy, clinical outcome (e.g., relapse, disease-specific survival, therapy failure), and/or clinical outcome as a function of time after diagnosis, time after initiation of therapy, and/or time after completion of treatment. Molecular co-variates can include, but are not limited to additional molecular marker values. For example, in embodiments wherein the FRα-expressing cancer is ovarian cancer, such markers may include, e.g., serum CA125 levels, serum DF3 levels, and/or plasma LPA levels.
  • In other aspects, the invention provides methods for monitoring the effectiveness of a therapy or treatment regimen. For example, the present invention provides methods for monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer. Specifically, the methods involve determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from said subject, wherein said subject has been previously administered MORAb-003; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein an increase or no change in the level of FRα in the sample derived from said subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is not efficacious; and wherein a decrease in the level of FRα in the sample derived from said subject as compared with the level of FRα in the control sample is an indication that the MORAb-003 treatment is efficacious.
  • For example, the control sample may be derived from a subject not subjected to the treatment regimen and a test sample may be derived from a subject subjected to at least a portion of the treatment regimen. Alternatively, the test sample and the control sample may be derived from the same subject. For example, the test sample may be a sample derived from a subject after administration of a therapeutic, such as MORAb-003. The control sample may be a sample derived from a subject prior to administration of therapeutic or at an earlier stage of therapeutic regimen. Accordingly, a decrease in the level of expression of FRα in the test sample, relative to the control sample, is an indication that therapy has decreased the progression of the FRα-expressing cancer, for example, ovarian cancer. For FRα-expressing cancers wherein a higher level of FRα is associated with a worse prognosis, such as e.g., ovarian cancer or squamous cell carcinoma of the head and neck, a decrease in the level of expression of FRα in the test sample, relative to the control sample, is an indication that therapy is effective in slowing the progression of the FRα-expressing cancer, or in causing a regression of the cancer, in the subject afflicted with the FRα-expressing cancer. In a preferred embodiment, the FRα-expressing cancer is ovarian cancer.
  • In various embodiments of this aspect of the invention, the sample may be urine, serum, plasma or ascites. In particular embodiments, the sample is urine or serum. Moreover, the FRα may be determined by contacting the sample with an antibody that binds FRα, optionally, using antibodies as described herein and assay methods as described herein.
  • In various embodiments, the MORAb-003 treatment antibody is (a) an antibody that comprises the heavy chain amino acid sequence as set forth in SEQ ID NO:7 and the light chain amino acid sequence as set forth in SEQ ID NO:8; (b) an antibody that binds the same epitope as the MORAb-003 antibody; or (c) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3..
  • In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In other embodiments, the FRα-expressing cancer is lung cancer. In more specific embodiments, the lung cancer is non-small cell lung cancer (NSCLC). In one such embodiment, the NSCLC is selected from the group consisting of adenocarcinoma, squamous cell lung carcinoma, large cell lung carcinoma, pleomorphic NSCLC, carcinoid tumor, salivary gland carcinoma, and unclassified carcinoma. In a preferred embodiment, the NSCLC is adenocarcinoma. In alternative embodiments, the lung cancer is small cell lung carcinoma (SCLC). In another embodiment, the lung cancer is bronchioalveolar carcinoma. In yet another embodiment, the lung cancer is a lung carcinoid tumor.
  • In another aspect, the invention provides methods of stratifying a subject with an FRα-expressing cancer into cancer therapy groups based on the determined level of FRα in a sample. In a preferred embodiment, the method involves stratifying a subject with an FRα-expressing cancer into one of at least four cancer therapy groups. In other embodiments, the method involves stratifying a subject with an FRα-expressing cancer into one of at least about two, about three, about four, about five, about six, about seven, about eight, about nine, or about ten cancer therapy groups.
  • According to the present invention, the levels of FRα may be associated with the severity, i.e., the stage, of the FRα expressing cancer. For example, ovarian cancer is stratified into different stages based on the severity of the cancer, as set forth herein. Accordingly, the present invention provides methods for stratifying ovarian cancer into Stage I, for example, Stage IA, Stage 1 B or Stage IC; Stage II, for example, Stage IIA, Stage IIB or Stage IIC; Stage III, for example, Stage IIIA, Stage IIIB or Stage IIIC; or Stage IV ovarian cancer.
  • SCLS or NSCLC may be stratified into different stages based on the severity of the cancer, as set forth herein. Accordingly, the present invention provides methods for stratifying the lung cancer, for example, SCLS or NSCLC, into the occult (hidden) stage; stage 0; Stage I, for example, stages IA and IB; Stage II, for example, stages IIA and IIB; Stage III, for example, stages IIIA and IIIB; or Stage IV lung cancer.
  • In yet another aspect, the present invention is predicated, at least in part, on the finding that FRα can serve as a predictive biomarker for treatment of FRα expressing cancers. Specifically, the methods of the present invention provide for assessing whether a subject will respond to treatment, for example, with MORAb-003, and whether and when to initiate treatment, for example, with MORAb-003, by assessing the levels of FRα in a subject.
  • In one aspect, the present invention provides a method for predicting whether a subject suffering from an FRα expressing cancer, for example, ovarian or lung cancer, will respond to treatment with MORAb-003, by determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from said subject; and comparing the level of folate receptor alpha (FRα) which is not bound to a cell in the sample derived from said subject with the level of FRα in a control sample, wherein a difference between the level of FRα in the sample derived from said subject and the level of FRα in the control sample is an indication that the subject will respond to treatment with MORAb-003.
  • In certain embodiments, the degree of difference between the levels of FRα not bound to a cancer cell in the test sample as compared to the control sample is indicative that the subject will respond to treatment with MORAb-003. For example, a difference of at least about 2-, about 3-, about 4-, about 5-, about 6-, about 7-, about 8-, about 9-, about 10-, about 15-, about 20-, about 25-, about 100-, about 500-, about 1000-fold or greater than the standard error of the assessment method is indicative that the subject will respond to treatment with MORAb-003. Alternatively or in combination, a difference of at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900% or about 1000% is indicative that the subject will respond to treatment with MORAb-003. Alternatively or in combination, a difference of at least about 1.5, and more preferably about two, about three, about four, about five or more standard deviations is indicative that the subject will respond to treatment with MORAb-003.
  • In various embodiments, the MORAb-003 treatment antibody is (a) an antibody that binds the same epitope as the MORAb-003 antibody; or (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3.
  • In various embodiments, the sample is urine, plasma, serum or ascites. In particular embodiments, the sample is urine or serum. In further embodiments, the FRα-expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer. In a particular embodiment, the FRα-expressing cancer is ovarian cancer. In another embodiment, the FRα-expressing cancer is non-small cell lung cancer, such as adenocarcinoma.
  • B. Anti-FRα Antibody Based Assays for Detecting FRα-Expressing Cancers
  • There are a variety of assay formats known to those of ordinary skill in the art for using an antibody to detect a polypeptide in a sample, including but not limited to enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunofluorimetry, immunoprecipitation, solution phase assay, equilibrium dialysis, immunodiffusion and other techniques. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988; Weir, D. M., Handbook of Experimental Immunology, 1986, Blackwell Scientific, Boston. For example, the assay may be performed in a Western blot format, wherein a protein preparation from the biological sample is submitted to gel electrophoresis, transferred to a suitable membrane and allowed to react with the antibody. The presence of the antibody on the membrane may then be detected using a suitable detection reagent, as is well known in the art and described below.
  • In another embodiment, the assay involves the use of an antibody immobilized on a solid support to bind to the target FRα polypeptide and remove it from the remainder of the sample. The bound FRα polypeptide may then be detected using a second antibody reactive with a distinct FRα polypeptide antigenic determinant, for example, a reagent that contains a detectable reporter moiety. As a non-limiting example, according to this embodiment the immobilized antibody and the second antibody which recognize distinct antigenic determinants may be any two of the monoclonal antibodies described herein selected from MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein. Alternatively, a competitive assay may be utilized, in which FRα is labeled with a detectable reporter moiety and allowed to bind to the immobilized anti-FRα antibody after incubation of the immobilized antibody with the sample. The extent to which components of the sample inhibit the binding of the labeled polypeptide to the antibody is indicative of the reactivity of the sample with the immobilized antibody, and as a result, indicative of the level of FRα in the sample.
  • The solid support may be any material known to those of ordinary skill in the art to which the antibody may be attached, such as a test well in a microtiter plate, a nitrocellulose filter or another suitable membrane. Alternatively, the support may be a bead or disc, such as glass, fiberglass, latex or a plastic such as polystyrene or polyvinylchloride. The antibody may be immobilized on the solid support using a variety of techniques known to those in the art, which are amply described in the patent and scientific literature.
  • In certain preferred embodiments, the assay for detection of FRα in a sample is a two-antibody sandwich assay. This assay may be performed by first contacting a FRα specific antibody (e.g., MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein) that has been immobilized on a solid support, commonly the well of a microtiter plate, with the biological sample, such that a soluble molecule naturally occurring in the sample and having an antigenic determinant that is reactive with the antibody is allowed to bind to the immobilized antibody (e.g., a 30 minute incubation time at room temperature is generally sufficient) to form an antigen-antibody complex or an immune complex. Unbound constituents of the sample are then removed from the immobilized immune complexes. Next, a second antibody specific for FRα is added, wherein the antigen combining site of the second antibody does not competitively inhibit binding of the antigen combining site of the immobilized first antibody to FRα (e.g., MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein, that is not the same as the monoclonal antibody immobilized on the solid support). The second antibody may be detectably labeled as provided herein, such that it may be directly detected. Alternatively, the second antibody may be indirectly detected through the use of a detectably labeled secondary (or "second stage") anti-antibody, or by using a specific detection reagent as provided herein. The subject invention method is not limited to any particular detection procedure, as those having familiarity with immunoassays will appreciate that there are numerous reagents and configurations for immunologically detecting a particular antigen (e.g., FRα) in a two-antibody sandwich immunoassay.
  • In certain preferred embodiments of the invention using the two-antibody sandwich assay described above, the first, immobilized antibody specific for FRα is a polyclonal antibody and the second antibody specific for FRα is a polyclonal antibody. In certain other embodiments of the invention, the first, immobilized antibody specific for FRα is a monoclonal antibody and the second antibody specific for FRα is a polyclonal antibody. In certain other embodiments of the invention the first, immobilized antibody specific for FRα is a polyclonal antibody and the second antibody specific for FRα is a monoclonal antibody. In certain other embodiments of the invention, the first, immobilized antibody specific for FRα is a monoclonal antibody and the second antibody specific for FRα is a monoclonal antibody. For example, in these embodiments it should be noted that monoclonal antibodies MORAb-003, MOV18, 548908, 6D398 or variants thereof as described herein, as provided herein recognize distinct and noncompetitive antigenic determinants (e.g., epitopes) on FRα polypeptides, such that any pairwise combination of these monoclonal antibodies may be employed. In other preferred embodiments of the invention, the first, immobilized antibody specific for FRα and/or the second antibody specific for FRα may be any of the kinds of antibodies known in the art and referred to herein, for example, by way of illustration and not limitation, Fab fragments, F(ab')2 fragments, immunoglobulin V-region fusion proteins or single chain antibodies. Those familiar with the art will appreciate that the present invention encompasses the use of other antibody forms, fragments, derivatives and the like in the methods disclosed and claimed herein.
  • In certain particularly preferred embodiments, the second antibody may contain a detectable reporter moiety or label such as an enzyme, dye, radionuclide, luminescent group, fluorescent group or biotin, or the like. The amount of the second antibody that remains bound to the solid support is then determined using a method appropriate for the specific detectable reporter moiety or label. For radioactive groups, scintillation counting or autoradiographic methods are generally appropriate. Antibody-enzyme conjugates may be prepared using a variety of coupling techniques (for review see, e.g., Scouten, W. H., Methods in Enzymology 135:30-65, 1987). Spectroscopic methods may be used to detect dyes (including, for example, colorimetric products of enzyme reactions), luminescent groups and fluorescent groups. Biotin may be detected using avidin or streptavidin, coupled to a different reporter group (commonly a radioactive or fluorescent group or an enzyme). Enzyme reporter groups may generally be detected by the addition of substrate (generally for a specific period of time), followed by spectroscopic, spectrophotometric or other analysis of the reaction products. Standards and standard additions may be used to determine the level of mesothelin polypeptide in a sample, using well known techniques.
  • A method of screening for the presence of an FRα expressing cancer according to the present invention may be further enhanced by the detection of more than one tumor associated marker in a biological sample from a subject. Accordingly, in certain embodiments the present invention provides a method of screening that, in addition to detecting reactivity of FRα not bound to a cell, also includes detection of at least one additional soluble marker of a malignant condition using established methods as known in the art and provided herein. As noted above, there are currently a number of soluble tumor associated antigens that are detectable in samples of readily obtained biological fluids.
  • C. Kits of the Invention
  • The invention also provides kits for assessing whether a subject is afflicted with an FRα-expressing cancer, for assessing the progression of an FRα-expressing cancer, for assessing the level of risk that a subject will develop an FRα-expressing cancer, or for monitoring the effectiveness of a therapy or treatment regimen for an FRα-expressing cancer. These kits include means for determining the level of expression of FRα and instructions for use of the kit to assess the progression of an FRα-expressing cancer, to assess the level of risk that a subject will develop an FRα-expressing cancer, or to monitor the effectiveness of a therapy or treatment regimen for an FRα-expressing cancer.
  • The kits of the invention may optionally comprise additional components useful for performing the methods of the invention. By way of example, the kits may comprise means for obtaining a sample from a subject, a control sample, e.g., a sample from a subject having slowly progressing cancer and/or a subject not having cancer, one or more sample compartments, and instructional material which describes performance of a method of the invention and tissue specific controls/standards.
  • The means for determining the level of FRα include known methods in the art for assessing protein levels, as discussed above, and specific preferred embodiments, for example, utilizing the MORAb-003 antibody, as discussed herein. Thus, for example, in one embodiment, the level of FRα is assessed by contacting a sample derived from a subject (such as urine or serum) with a folate receptor alpha (FRα) binding agent. In a preferred embodiment, the binding agent is an antibody. Many of the types of antibodies that bind FRα are discussed above in the methods of the invention and may also be utilized in the kits of the invention.
  • The means for determining the level of FRα can further include, for example, buffers or other reagents for use in an assay for determining the level of FRα. The instructions can be, for example, printed instructions for performing the assay and/or instructions for evaluating the level of expression of FRα.
  • The kits of the inventions may also include means for isolating a sample from a subject. These means can comprise one or more items of equipment or reagents that can be used to obtain a fluid or tissue from a subject. The means for obtaining a sample from a subject may also comprise means for isolating blood components, such as serum, from a blood sample. Preferably, the kit is designed for use with a human subject.
  • III. Screening Assays
  • In further embodiments, the invention also provides methods (also referred to herein as "screening assays") for identifying modulators, i.e., candidate or test compounds or agents (e.g., proteins, peptides, peptidomimetics, peptoids, small molecules or other drugs), which modulate the growth, progression and/or aggressiveness of cancer, e.g., an FRα-expressing cancer, or a cancer cell, e.g., an ovarian cancer cell, by monitoring and comparing the levels of FRα in a sample. Such assays typically comprise a test compound, or a combination of test compounds, whose activity against cancer or a cancer cell is to be evaluated. Compounds identified via assays such as those described herein may be useful, for example, for modulating, e.g., inhibiting, ameliorating, treating, or preventing aggressiveness of an FRα-expressing cancer or a cancer cell, e.g., an ovarian cancer cell. By monitoring the level of FRα in a sample, one can determine whether the FRα-expressing cancer is progressing or regressing and whether the test compound has the desired effect. For example, in embodiments wherein the FRα-expressing cancer is a cancer for which higher levels of FRα are associated with a worse prognosis, a decrease in the level of FRα after administration of the test compound(s) would be indicative of the efficacy of the test compound. By contrast, an increase in the level of FRα after administration of the test compound(s) would indicate that the test compound is not effective in treating ovarian cancer. By contrast, in embodiments wherein the FRα-expressing cancer is a cancer for which higher levels of FRα are associated with a better prognosis, an increase in the level of FRα after administration of the test compound(s) would be indicative of the efficacy of the test compound. By contrast, an decrease in the level of FRα after administration of the test compound(s) would indicate that the test compound is not effective in treating ovarian cancer.
  • The test compounds used in the screening assays of the present invention may be obtained from any available source, including systematic libraries of natural and/or synthetic compounds. Test compounds may also be obtained by any of the numerous approaches in combinatorial library methods known in the art, including biological libraries; peptoid libraries (libraries of molecules having the functionalities of peptides, but with a novel, non-peptide backbone which are resistant to enzymatic degradation but which nevertheless remain bioactive; see, e.g., Zuckermann et al., 1994, J. Med. Chem. 37:2678-85); spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound' library method; and synthetic library methods using affinity chromatography selection. The biological library and peptoid library approaches are limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, 1997, Anticancer Drug Des. 12:145).
  • Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90:6909; Erb et al. (1994) Proc. Natl. Acad. Sci. USA 91:11422; Zuckermann et al. (1994). J. Med. Chem. 37:2678; Cho et al. (1993) Science 261:1303; Carrell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2059; Carell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2061; and in Gallop et al. (1994) J. Med. Chem. 37:1233.
  • Libraries of compounds may be presented in solution (e.g., Houghten, 1992, Biotechniques 13:412-421), or on beads (Lam, 1991, Nature 354:82-84), chips (Fodor, 1993, Nature 364:555-556), bacteria and/or spores, ( Ladner, USP 5,223,409 ), plasmids (Cull et al, 1992, Proc Natl Acad Sci USA 89:1865-1869) or on phage (Scott and Smith, 1990, Science 249:386-390; Devlin, 1990, Science 249:404-406; Cwirla et al, 1990, Proc. Natl. Acad. Sci. 87:6378-6382; Felici, 1991, J. Mol. Biol. 222:301-310; Ladner, supra.).
  • The present invention is further illustrated by the following examples which should not be construed as further limiting. The contents of all references, patents and published patent applications cited throughout this application, as well as the Figures, are expressly incorporated herein by reference in their entirety.
  • EXAMPLES EXAMPLE 1. DETERMINATION OF FRα LEVELS IN URINE SAMPLES FROM HUMAN SUBJECTS WITH AND WITHOUT OVARIAN CANCER AS MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA). Materials and Methods
  • Urine samples were obtained from human subjects, including subjects afflicted with ovarian cancer and normal control subjects not afflicted with ovarian cancer. The levels of FRα in urine samples were determined using an electrochemiluminescence immunoassay (ECLIA) according to the following procedure (see Namba et al. (1999) Analytical Science 15:1087-1093):
  • i. Antibody coating to micro beads
  • The monoclonal anti-folate receptor alpha antibody was coated over the surface of micro beads (Dynabeads M-450 Epoxy, Dynal). Thirty six milligrams of micro beads were mixed with 1.2 mL of antibody MOV18 (0.36 mg/mL, Enzo Life Science) in 0.15 mol/L phosphate buffer saline pH 7.8 (PBS), followed by gentle mixing for 16 hours at room temperature. The micro beads were then washed 5 times with 50 mM HEPES buffer containing 0.1 % normal rabbit serum (NRS), 150 mmol/L NaCl, 0.01 % Tween 20 pH 7.5 (wash buffer). Thereafter, the coated micro beads were suspended in 1.2 mL 50 mM HEPES buffer containing 20% NRS, 150 mmol/L NaCl and 0.01% Tween 20 pH 7.5 (reaction buffer) to block the unbound surface, followed by gentle mixing for 3.5 hours at room temperature. Finally, the micro beads were washed 5 times with wash buffer and re-suspended with 1.2 mL 50 mM HEPES buffer containing 10% NRS, 150 mmol/L NaCl, 10 mmol/L EDTA-2Na and 0.01% Tween 20 pH 7.5 (reaction buffer) so that the concentration of micro beads was 30 mg/mL. The micro beads were stored at 4°C until use.
  • ii. Antibody labeling with Ruthenium-chelate-NHS (Ru)
  • One milliliter of MORAB-003 (1 mg/mL) in PBS was mixed with 14 µL of Ru (10 mg/mL), initial molar ratio of antibody to Ru was 1:20, followed by shaking for 30 minutes at room temperature in the dark. The reaction was terminated by adding 25 µL of 2 mol/L glycine solution followed by incubation for 20 minutes. The labeled antibody was purified by gel filtration using Sephadex G-25 (GE Healthcare) eluted with PBS. The first eluted yellow fraction was collected and the concentration of antibody and Ru were determined by means of the Pierce BCA protein assay kit (Thermo Scientific) and absorption at 455 nm respectively. The final molar ratio was calculated by the formula: Final molar ratio = [(absorption at 455)/13700]/[Ab(mg/mL/150000)]. The labeled antibody was stored at 4°C until use.
  • iii. One Step Immunoassay
  • The antibody coated micro beads were set on the reagent table of the Picolumi 8220 (Sanko, Tokyo, Japan) after adjusting the concentration of the beads to 1.5 mg/ml (working solution) in reaction buffer. The Ru labeled antibody was set on the reagent table of the Picolumi 8220 after adjusting the concentration of antibody to 2 µg/ml (working solution) in reaction buffer.
  • Ten microliters of urine (diluted 1:51 in reaction buffer) or standard FRα (prepared in reaction buffer) and 100 µL of reaction buffer were dispensed into a reaction tube (Sanko, Tokyo, Japan) and set on Picolumi 8220.
  • The following steps were automatically run by Picolumi 8220. Twenty five microliters of beads (working solution) and 180 µL of Ru labeled antibody (working solution) were dispensed. After 26 minutes of incubation at 30 +/-2°C, the beads were washed and suspended with 300 µL of electrolyte solution (Sanko, Tokyo, Japan). The washed beads were subsequently transferred to the electrode and electrochemiluminescence (ECL) emission was measured.
  • All ECL measurements were carried out in duplicate.
  • Results
  • Table 1 depicts the urine levels of FRα in individual subjects with ovarian cancer and non-afflicted female control subjects. Table 1: FRα levels in urine of subjects with ovarian cancer and normal female control subjects
    Group Sample # FRα (pg/mL)
    ovarian cancer 1 27800
    2 40242
    3 85580
    4 4994
    5 2017
    6 3781
    7 29469
    8 47456
    9 4479
    10 11920
    11 18352
    12 162017
    13 30630
    14 14431
    15 11801
    16 13470
    17 11563
    18 22185
    19 52106
    normal control 20 8491
    21 4885
    22 3595
    23 21301
    24 22757
    25 16578
    26 6081
    27 4195
    28 12169
    29 20639
  • Figure 2 depicts the distribution of FRα levels in urine in subjects with ovarian cancer and in normal female control subjects, as set forth in Table 1.
  • Table 2 summarizes the number of subjects (n), mean, standard deviation (SD), maximum (Max.) and minimum (Min.) values for the levels of FRα in the ovarian cancer group and the normal female control group. Table 2: Summary of urine FRα measurement
    FRα (pg/mL)
    ovarian cancer normal female
    N 19 10
    Mean 31279 12069
    SD 37895 7654
    Max. 162017 22757
    Min. 2017 3595
  • Discussion
  • A high level of FRα was detected in the urine of subjects with ovarian cancer. Moreover, the levels of FRα differed significantly between ovarian cancer and normal female control groups (p=0.03, one-sided).
  • EXAMPLE 2. DILUTION LINEARITY - DETERMINATION OF FRα LEVELS IN SERIALLY DILUTED URINE SAMPLES MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA)
  • Dilution Linearity is a measure of accuracy of an assay. Two urine samples were serially diluted by a factor of 10 and 100. The FRα levels of each sample were measured as set forth in Example 1 and compared to assess the percent error. Percent error was calculated as follows: FRα in undiluted sample * dilution factor FRα in undiluted sample * 100 FRα in undiluted sample
    Figure imgb0001
    The results are set forth in Table 3: Table 3: Dilution Linearity for Urine
    Sample Dilution factor FRα (pg/mL) Error (%)
    1 1 25037 -
    10 2601 4
    100 279 11
    2 1 16649 -
    10 1696 2
    100 173 4
    The foregoing results demonstrate dilution linearity in assessing the levels of FRα in human urine samples and that, within acceptable errors, urine can be diluted up to a factor of at least 100 while retaining accurate levels of FRα. Accordingly, dilution of the urine samples may be considered prior to determining levels of FRα.
  • EXAMPLE 3: CENTRIFUGATION OF URINE SAMPLES - ADDRESSING REPRODUCIBILITY
  • The reproducibility of the ECLIA assay for a particular urine sample was also tested. For example, as reflected in Table 4, ECLIA assays of the same sample resulted in varying results. Table 4. Reproducibility without sample centrifugation
    ECL Counts
    Sample Test
    1 Test 2
    1 29380 15046
    2 20912 17227
  • The presence of insoluble material (precipitates) in urine samples was hypothesized to be responsible for the variability seen in measuring the levels of FRα. As a result, centrifugation of samples in order to remove urine sediment, prior to measurement of FRα levels, was considered as an option to enhance the accuracy and reproducibility of the assay.
  • Table 5 depicts the results obtained when three samples were centrifuged prior to performance of the ECLIA assay. Table 5. Reproducibility with sample centrifugation
    FRα concentration (ng/mL)
    Test Sample 1 Sample 2 Sample 3
    1 10.4 9.2 13.3
    2 10.5 8.9 14.0
    3 10.3 9.2 13.4
    Mean 10.4 9.1 13.6
    SD 0.1 0.1 0.3
    CV(%) 1.0 1.1 2.2
    As set forth above, the results indicate that centrifugation provided more consistent measurements of FRα concentration.
  • Additionally, two samples were subjected to (i) centrifugation (at 2000 x g for 2 min) and the supernatant removed for measurement of FRα (depicted as sample "A" below in Table 6) and (ii) centrifugation followed by vortexing (depicted as sample "B" below in Table 6), prior to measurement of FRα levels by the ECLIA assay set forth in Example 1. The results are reflected in Table 6 below. Table 6: Effect of centrifugation on FRα levels in urine
    Sample Sediment after centrifugation A/B FRα (pg/mL) Difference (%)
    1 Yes (++) A 13678 -
    B 16559 21
    2 Yes (+) A 12271 -
    B 13206 8
  • Difference (%) was determined as follows: Level of FRα in ʺBʺ Level of FRα in ʺAʺ * 100 Level of FRα in ʺAʺ
    Figure imgb0002
  • As shown in Table 6, the levels of FRα as determined by the ECLIA assay vary depending on whether urine was clarified by centrifugation to remove precipitates or whether urine was vortexed to suspend or disperse sediments. Accordingly, in certain embodiments centrifuging or vortexing of urine samples may be performed prior to determining levels of FRα.
  • EXAMPLE 4. DETERMINATION OF FRα LEVELS IN CENTRIFUGED URINE SAMPLES FROM HUMAN SUBJECTS WITH AND WITHOUT OVARIAN CANCER MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA).
  • Based on the results of Example 3, the assay for assessing FRα levels in subjects was modified to introduce a centrifugation step. FRα levels were determined on the same samples utilized in Example 1, including the group of subjects with ovarian cancer and the group of normal female control subjects.
  • Materials and Methods
  • The methodology utilized was as described in Example 1, except that the urine samples were centrifuged for 10000 x g for 1 minute and the resulting supernatant subsequently diluted by 1:51 in reaction buffer.
  • Results
  • Table 7 depicts the levels of FRα in centrifuged and non-centrifuged urine samples from subjects afflicted with ovarian cancer and healthy female control subjects. Table 7: Urine FRα1 level in ovarian cancer and normal control group
    FRα (pg/mL)
    Group Sample # Without centrifugation With centrifugation sediment after centrifugation
    ovarian cancer 1 27800 23960 +
    2 40242 37852 +
    3 85580 78976 +
    4 4994 3766 +
    5 2017 1512 -
    6 3781 3443 -
    7 29469 25728 +
    8 47456 16556 +
    9 4479 3357 -
    10 11920 5020 +
    11 18352 16695 -
    12 162017 82705 +
    13 30630 4496 +
    14 14431 8786 +
    15 11801 10582 -
    16 13470 5611 +
    17 11563 5463 +
    18 22185 14443 +
    19 52106 38327 -
    normal control 20 8491 6867 +
    21 4885 3754 -
    22 3595 3529 -
    23 21301 15047 +
    24 22757 4850 +
    25 16578 14366 +
    26 6081 5201 -
    27 4195 3135 -
    28 12169 499 +
    29 20639 2439 +
  • According to the results set forth in Table 7, centrifugation resulted in a decrease in the measurement of FRα levels in some samples, as previously demonstrated in Table 6.
  • EXAMPLE 5: DETECTION OF FRα IN URINE SEDIMENT BY IMMUNOBLOTTING
  • Based on the results shown in Examples 3 and 4, the presence or absence of FRα in urine sediment/precipitate was assessed using western blotting.
  • Materials and Methods
  • Urine samples from 2 ovarian cancer patients for whom FRα concentrations were measured at 18,747 pg/mL and 145,564 pg/mL, respectively (See Table 10 supra), were subjected to the following procedures. Control samples consisted of HeLa cell lysate 10 µg, liver tissue lysate 20 µg, and ovarian cancer tissue lysate 20 µg.
    • 900 µL urine was centrifuged for 2 minutes at 10000g
    • supernatant was removed
    • the remaining pellet was dissolved in 15 µL of PAGE sample buffer (containing 292 mM LDS) and subsequently boiled at 70°C for 10 min
    • The entire sample (approx. 20 µL) was loaded onto the NuPAGE bis-tris gel (Invitrogen)
    • After electrophoresis, proteins were transferred to PVDF membrane
    • 1% skim milk / 0.05% Tween 20 / PBS was added for blocking
    • The membrane was washed with 0.05% Tween 20 / PBS
    • 0.5 mL of monoclonal antibody 548908 (R&D Systems) at 2 µg/ml was added and incubated for 60 minutes at room temperature
    • The membrane was washed with 0.05% Tween 20 / PBS
    • 10 mL of anti-mouse IgG-HRP (DAKO p0447, 1:2000) was added and allowed to incubate for 60 minutes
    • The membrane was washed with 0.05% Tween 20 / PBS
    • Pierce ECL Substrate was added to the membrane
    • The membrane was removed from the substrates and then imaged using the LAS-3000 (FUJIFILM) system
    Results
  • The resulting immunoblot is shown in Figure 3. In this figure, lanes 1-5 correspond to FRα detected from the following sources:
    1. (1) urine from ovarian cancer patient with a measured FRα level of 18,747 pg/mL
    2. (2) urine from ovarian cancer patient with a measured FRα level of 145,564 pg/mL
    3. (3) HeLa cell lysate: 10 µg
    4. (4) Liver tissue lysate: 20 µg
    5. (5) ovarian cancer tissue lysate: 20 µg
  • Lane 6 in the western blot represents molecular weight markers and demonstrates that the observed band in lanes 1, 2, 3 and 5 runs at the expected molecular weight for FRα.
  • Lanes 3 and 5 are positive control samples and lane 4 is a negative control sample. The faint band on lane 1 and the clear band on lane 2 demonstrate that FRα can be detected in the urine sediment of ovarian cancer patients by western blotting.
  • EXAMPLE 6. DETERMINATION OF FROG LEVELS IN GUANIDINE-TREATED NORMAL HUMAN URINE SAMPLES BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA).
  • Based on the results of Example 4 in which centrifugation resulted in decreased FRα levels, and the results of Example 5 where the urine sediment obtained from centrifugation was shown to contain immunoreactive FRα, methods were sought to solubilize the sediments of urine to obtain more quantitative and accurate measurements of FRα.
  • In this regard, treatment of normal female urine samples with guanidine prior to assessing FRα levels was attempted.
  • The methodology utilized was as described in Example 1, except that the urine samples were mixed in a 1:1 ratio with either 6 M guanidine in buffer (PBS) or buffer alone. Subsequently, the urine samples were diluted by 1:51 in reaction buffer.
  • The results of this assay are shown in Table 8. Table 8: Normal urine FRα level with or without guanidine treatment
    Sample Guanidine treatment FRα (pg/mL)
    Std Ag Yes 83964
    No 82512
    Normal Urine 1 Yes 9431
    No 7796
    2 Yes 5713
    No 4066
    3 Yes 9687
    No 9428
    The results of this experiment indicate that guanidine does not interfere with FRα measurements. As can be seen for the pure antigen control (Std Ag), this methodology of guanidine treatment and subsequent dilution has no effect on the measurement of FRα. Further, it will be noted that in all three (3) urine samples assessed, the levels of FRα were higher in the samples treated (solubilized) with guanidine relative to the samples not treated with guanidine.
  • The reliability of guanidine pre-treatment of urine samples was further assessed by exposing three samples to guanidine and measuring the FRα concentration of each guanidine treated sample 3 times using the ECLIA assay. The results are reflected in Table 9 below: Table 9: Intra-assay reproducibility of guanidine treated urine
    FRα (pg/mL)
    Test Sample 1 Sample 2 Sample 3
    1 9210 5477 9889
    2 9638 5405 10047
    3 10192 5812 10944
    Mean 9680 5565 10293
    SD 492 217 569
    CV(%) 5.1 3.9 5.5
    As set forth above, the results indicate that guanidine treatment of urine prior to FRα assay provided consistent measurements of FRα concentration with very low CV's.
  • EXAMPLE 7. DETERMINATION OF FRα LEVELS IN GUANIDINE-TREATED URINE SAMPLES FROM HUMAN SUBJECTS WITH AND WITHOUT OVARIAN CANCER MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA).
  • Based on the results of Example 6 in which guanidine treatment was shown not to interfere with FRα assays, a modified assay protocol was employed to measure FRα in the urine samples from the subjects with and without ovarian cancer in Example 1.
  • The following assay protocol was employed:
  • Materials and Methods
  • The methodology utilized was as described in Example 1, except that the urine samples were mixed in a 1:1 ratio with a 6 M guanidine buffer and subsequently diluted by 1:26 in reaction buffer.
  • Results
  • Table 10 depicts the levels of FRα in guanidine treated urine samples from subjects afflicted with ovarian cancer and healthy female control subjects. Table 10: Urine FRα level in ovarian cancer and normal control group
    Group Sample # FRα (pg/mL)
    ovarian 1 27015
    cancer 2 37315
    3 79579
    4 285
    5 1864
    6 2902
    7 27914
    8 51864
    9 2699
    10 9455
    11 18396
    12 145564
    13 19046
    14 10440
    15 10977
    16 9199
    17 18223
    18 18747
    19 51098
    normal control 20 8012
    21 3797
    22 3323
    23 20976
    24 6941
    25 14512
    26 7286
    27 2789
    28 2617
    29 7233
    Figure 4 shows the distribution of FRα levels in the urine of ovarian cancer afflicted subjects and normal female control subjects using the modified protocol with guanidine treatment. A statistically significant difference between groups was observed. Table 11 1 summarizes these results. Table 11: Summary of urine FRα measurement
    FRα (pg/mL)
    ovarian cancer normal control
    n 19 10
    Mean 28557 7749
    SD 34990 5850
    Max. 145564 20976
    Min. 285 2617
  • Using the data from this experiment, a receiver operating characteristic (ROC) analysis was performed. Figure 5 shows an ROC curve of the sensitivity and specificity of the ECLIA measurement of FRα levels in urine after the urine was treated with guanidine. AUC is the area under the curve, which measures the accuracy of the test in separating ovarian cancer from control subjects.
  • Using an arbitrary cutoff value of 9100 pg FRα/mL, the AUC was 0.70 with a positive predictive value of 70% and a negative predictive value of 80%, as shown in Table 12. Using this cutoff value, 15/19 ovarian cancer patients had a concentration of FRα above 9100 pg/mL and 8/10 normal subjects had a concentration of FRα less than 9100 pg/mL. Table 12: Guanidine treatment for urine measurement
    ovarian cancer control
    Number of Samples 19 10
    Positive 15 2
    Predictive value (%) 78.9 80.0
  • EXAMPLE 8: CREATININE CORRECTION OF FRα CONCENTRATIONS DETERMINED IN GUANIDINE-TREATED URINE SAMPLES BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA)
  • Concentrations of FRα were previously determined using ECLIA of guanidine-treated urine samples from ovarian cancer patients and normal female controls (See Example 7, Table 10). Here, these FRα concentrations were corrected for urine creatinine levels in order to normalize for the glomerular filtration rate. The resulting values were subjected to an ROC analysis.
  • Methods
  • The urine creatinine level was determined by the Ministry of Health, Labour and Welfare approved test kit, determiner L CRE (Kyowa Medex, Japan). The corrected value for urine FRα concentration was calculated as follows: Urine FRα Creatinine correction ng / g = Urine FRα ng / L × 1000 / Urine Creatinine mg / dL × 10 = Urine FRα ng / L × 1000 / Urine Creatinine mg / L = Urine FRα ng / L / Urine Creatinine g / L = Urine FRα ng / Urine Creatinine g or = 1 / 1000 × Urine FRα μg / Urine Creatinine g
    Figure imgb0003
  • Results
  • Table 13 presents the resulting creatinine-corrected FRα levels. Table 13: Creatinine-corrected FRα levels determined using ECLIA of guanidine-treated urine samples
    Group Sample # FRα (pg/mL) Corrected FRα (µg FRα/ g creatinine)
    ovarian cancer 1 27015 11.6
    2 37315 37.8
    3 79579 33.9
    4 285 0.6
    5 1864 6.7
    6 2902 7.1
    7 27914 54.9
    8 51864 17.1
    9 2699 13.5
    9 9455 14.5
    10 18396 23.1
    11 145564 66.0
    12 19046 9.1
    13 10440 8.5
    14 10977 7.4
    15 9199 5.9
    16 18223 13.0
    17 18747 9.6
    normal control 18 3797 7.7
    19 3323 7.9
    20 20976 10.7
    21 6941 4.3
    22 14512 8.6
    23 7286 13.8
    24 2789 4.3
    25 2617 1.9
    26 7233 3.1
  • Figure 6 shows the distribution of FRα levels in ovarian cancer (OC) and normal female control subjects after correction for urine creatinine levels. There is a statistically significant difference between ovarian cancer patients and controls in creatinine-corrected levels of FRα (p=0.007).
  • The summary data for ovarian cancer and normal control subjects are provided in Table 14. Table 14: Summary statistics for creatinine-corrected FRα levels
    FRα (µg/g-creatinine)
    ovarian cancer normal control
    n
    18 9
    Mean 18.9 6.9
    SD 17.9 3.9
    Max. 66.0 13.8
    Min. 0.6 1.9
  • The creatinine-corrected FRα levels were further subjected to an ROC analysis. The ROC curve is shown in Figure 7. Table 15 presents the sensitivity, specificity, and area under the curve (AUC) for various cutoff values of the creatinine-corrected test. Table 15: Sensitivity, specificity, and AUC for various cutoff values of the creatinine-corrected FRα test
    Cut-off Sensitivity Specificity AUC
    3.0 94.4% 11.1% 0.67
    4.0 94.4% 22.2% 0.70
    5.0 94.4% 44.4% 0.78
    6.0 88.9% 44.4% 0.74
    9.0 66.7% 77.8% 0.70
  • As previously noted, there is a clear discrimination between urines of ovarian cancer patients and those from healthy female control subjects.
  • EXAMPLE 9: ENZYME IMMUNOASSAY (EIA) AND OPTIMIZATION THEREOF 1. Enzyme Immunoassay (EIA) Antibody coating to microtiter plates
  • The monoclonal anti-folate receptor alpha antibody was coated on the surface of microtiter plates (Nunc-immunoplate, Thermo Scientific) as follows. One hundred microliter of antibody (absorbance 0.02 at 280 nm) in 50 mmol/L carbonate buffer pH 9.4 was dispensed into wells, followed by coating for 16 hours at 4°C. The microplates were then washed 2 times with PBS containing 0.05% Tween20 (PBS-T). Thereafter 0.15 mL of PBS containing 20% normal rabbit serum pH 7.8 was dispensed into wells to block the unbounded surface, followed by blocking for 1 hour at room temperature. Finally, the microplates were washed 2 times with PBS-T. The antibody coated plates were dried and kept at 4°C in aluminum bags until use.
  • Biotin labeling
  • Biotin labeling was conducted according to the manufacturers recommendations for the EZ-Link Sulfo-NHS-LC-LC-Biotin (Product No. 21338, Thermo Scientific). Briefly, 1 mg of antibody in 0.4 mL of PBS was mixed with 0.013 mL of 10 mM Sulfo-NHS-LC-LC-Biotin, with an initial molar ratio of antibody to biotin of 1:20, followed by incubation for 30 min at room temperature. The biotin coupled antibody was purified by gel filtration using a PD-10 column (GE Healthcare) eluted with PBS to remove non-reacted biotin. In order to determine the level of biotin incorporation, the EZ Biotin quantitation kit (Product No. 28005, Thermo Scientific) was used. The biotin labeled antibody was stored at -80°C until use.
  • Two step immunoassay
  • For the first reaction, 40 µL of plasma or standard antigen and 60 µL of 50 mM HEPES buffer containing 10% NRS, 150 mmol/L NaCl, 10 mmol/L EDTA-2Na, 0.01% Tween 20 pH 7.5 (reaction buffer) was dispensed into antibody coated wells. The plate was incubated for 18 hours at 4°C, and subsequently washed 5 times with PBS-T. For the second reaction, 100 µL of 10 µg/mL biotin labeled antibody in reaction buffer was dispensed. The plate was incubated for 1 hour at room temperature, and subsequently washed 5 times with PBS-T. 100 µL of horse radish peroxidase labeled streptavidin (Pierce) was dispensed. After 30 minutes incubation at room temperature, the plates were washed 5 times with PBS-T. Finally, for the color development, 100 µL of TMB solution (KPL) was dispensed and left for 15 minutes in dark. After stopping color development by adding 100 µL of 1N HCl, the absorption at 450 nm was read using a plate reader. All washing steps were automatically done by auto-plate washer (AMW-8, BioTec, Japan), and all EIA measurements were carried out in duplicate.
  • Figure 8 depicts the EIA assay using MOV18 as the capture antibody and biotinylated MORAb-003 as the detector antibody.
  • 2. Optimization of EIA procedures
  • The above EIA procedures were arrived at in part based on the following experiments designed to optimize the procedure.
  • First, avidin-HRP, biotin labeled antibody and HRP labeled antibody were compared. Compared with HRP labeled antibody, biotin labeled antibody and avidin-HRP provided a higher signal; therefore, biotin labeled antibody and avidin-HRP were employed.
  • Second, one and two-step incubation procedures were compared. As depicted in Figure 9, a two-step incubation procedure yielded a higher signal and was thus employed.
  • Third, to optimize the second incubation time, incubation times of one to four hours were compared. The results indicated that one hour incubation times provided the highest signal to noise ratio and therefore an incubation time of one hour was subsequently employed.
  • Fourth, in order to optimize the working concentration of biotin labeled antibody, HRP labeled antibody and sample volume, various concentrations were employed as set forth in the above description of the EIA assay. The optimal values concentrations are described above.
  • EXAMPLE 10: COMPARISON OF FRα IN HUMAN PLASMA USING ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA) AND ENZYME IMMUNOASSAY (EIA)
  • The levels of FRα were measured in human plasma samples taken from ovarian cancer patients and healthy female controls using the electrochemiluminescence assay (ECLIA) described in Example 1 and Figure 1 (using MORAb-003 as the capture antibody and ruthenium (Ru)-labeled MOV-18 as the labeled detector antibody) and the enzyme immunoassay (EIA) described in Example 9 and Figure 8. In both assays, 40 µL of plasma was assayed.
  • Table 16 shows the plasma levels of FRα in ovarian cancer and normal control subjects, as determined using the EIA and ECLIA. Table 16: Plasma concentrations of FRα determined using EIA and ECLIA methods
    Group Sample # FRα concentration (pg/mL)
    EIA ECLIA
    Ovarian cancer 1 10 73
    2 <10 200
    3 56 286
    4 44 286
    5 353 1606
    6 83 494
    Healthy control 7 110 127
    8 162 112
    9 88 252
    10 180 254
    11 262 471
    12 206 396
  • With only one exception, the results for all of the subjects indicated that the concentrations of FRα detected in serum using EIA are lower than the levels detected using ECLIA, demonstrating that the EIA assay, as formatted, is not as sensitive as the ECLIA assay when this particular combination of capture (MOV-18) and detector antibodies (MORAb-003) is used. Therefore, further experiments with other types of antibodies were conducted to develop a more sensitive EIA procedure.
  • EXAMPLE 11: FEASIBILITY OF DIFFERENT TYPES OF ANTIBODIES FOR EIA MEASUREMENT OF FROG IN HUMAN PLASMA 1, Preliminary experimentation of Antibody Combinations
  • Various combinations of capture/detector antibodies were considered. Preliminary experimentation rendered the results set forth in Table 17.
    Figure imgb0004
  • 2. Comparison of EIA assays using various antibody combinations and comparison to the ECLIA assay
  • The levels of FRα in plasma from ovarian cancer patients and normal healthy female controls were measured using an enzyme-linked immunosorbent assay (EIA) with different combinations of capture and biotin-labeled antibodies and compared with the levels of FRα measured using the ECLIA assay.
  • Materials and Methods
  • The ECLIA method was as described in Example 1 and depicted in Figure 1 (using the MORAb-003 antibody as the capture antibody and the Mov-18 antibody as the labeled detector antibody). The EIA method was as described in Example 9, except that three different combinations of capture/detector antibodies were employed, as depicted in Figure 10: MOV18/MORAb-003, 548908/MORAb-003 and 6D398/MORAb-003. The antibodies 548908 and 6D398 are commercially available. The 548908 antibody was obtained from R&D Systems (North Las Vegas, NV) and the 6D398 antibody was obtained from US Biological (Swampscott, MA 01907).
  • Results
  • The concentrations of FRα (pg/mL) determined using the EIA and ECLIA methods are shown in Table 18. In addition, the concentrations of FRα (pg/ml) determined by EIA using various combinations of capture/detector antibodies are depicted graphically in Figure 11. Table 18: Plasma concentrations of FRα (pg/mL) determined using the EIA and ECLIA methods with various combinations of capture and detector antibodies.
    Group Sample # EIA 548908-MORAb-003 EIA 6D398-MORAb-003 EIA MOV18-MORAb-003 ECLIA MOV18-MORAb-003
    Ovarian cancer 1 176 2 10 217
    2 85 <0 - 165
    3 257 35 - 296
    4 117 42 44 322
    5 2048 370 353 1335
    6 447 63 83 390
    Normal control 7 247 66 110 137
    8 213 110 162 185
    9 367 78 88 219
    10 364 152 180 228
    11 804 224 262 388
    12 473 194 206 318
  • The data in Table 18 indicate that the measurements of FRα levels with EIA using the 54908-MORAb-003 combination yields results that are most similar to the results obtained using the ECLIA assay. Quantitive analyses were performed, confirming this observation. Further, these data demonstrate that the detection of FRα is highly dependent on the antibodies and antibody combination employed. Accordingly, different antibody combinations can be employed for the determination of FRα in biological fluids. In addition, since the data obtained from the EIA and the ECLIA assay formats are similar, various assay formats can be used for the determination of FRα.
  • For each of the three combinations of capture and detector antibodies used for the EIA method, a regression analysis was performed, and the concentrations of FRα (pg/mL) determined with EIA were correlated with the concentrations determined with the ECLIA assay. The results of this analysis are shown in Table 19 and in Figure 12. Table 19: Correlations of plasma concentrations of FRα measured by ECLIA with concentrations measured by EIA using three combinations of capture and detector antibodies
    Capture antibody-detector antibody 548098-MORAb-003 6D398-MORAb-003 MOV18-MORAB-003
    r 0.960 0.781 0.715
    Slope 1.595 0.285 0.223
    Intercept -87.06 -0.64 58.62
    The results for EIA using the 548098-MORAb-003 capture-detector combination correlated highly (r=0.96) with the results for ECLIA.
  • EXAMPLE 12: PLASMA LEVELS OF FRα DETERMINED By EIA AND ECLIA IN SAMPLES FROM OVARIAN CANCER PATIENTS
  • Measurements of serum FRα levels were determined in a group of ovarian cancer patients (n=17) and normal controls (n=35) using ECLIA and EIA. For the EIA measurements, the 548908 capture/ MORAb-003 detector antibody combination was employed. The EIA procedure was otherwise as described in Example 9. The ECLIA procedure was as described in Example 1. The results are shown in Table 20. Table 20: Plasma FRα concentrations in ovarian cancer patients and normal controls, as determined using EIA and ECLIA
    Group Sample # EIA ECLIA
    pg/mL pg/mL
    Ovarian cancer
    1 245 217
    2 247 223
    3 194 229
    4 2613 1335
    5 154 153
    6 319 215
    7 516 390
    8 370 271
    9 933 449
    10 4768 4502
    11 385 266
    12 251 322
    13 404 349
    14 338 371
    15 4147 2344
    16 179 165
    17 380 296
    Control 18 232 181
    19 372 173
    20 332 189
    21 380 203
    22 376 290
    23 406 217
    24 281 182
    25 348 191
    26 490 247
    27 253 137
    28 368 185
    29 338 195
    30 289 219
    31 338 206
    32 406 226
    33 365 228
    34 501 280
    35 806 388
    36 613 286
    37 380 250
    38 420 281
    39 393 280
    40 552 284
    41 664 318
    42 429 261
    43 499 286
    44 310 218
    45 281 217
    46 215 202
    47 293 217
    48 380 256
    49 270 195
    50 393 234
    51 425 308
    52 226 199
  • Figure 13 shows the distribution of plasma FRα concentrations in subjects with ovarian cancer and normal female control subjects as determined using EIA.
  • Table 21 shows summary descriptive statistics for the plasma FRα concentrations in ovarian cancer and normal female control subjects as determined using EIA. Table 21: Summary of FRα plasma concentrations in ovarian cancer and normal female control subjects as determined using EIA.
    FRα (pg/mL)
    Ovarian cancer Normal control
    n 17 35
    Mean 967 389
    SD 1438 126
    Max. 4768 806
    Min. 154 215
  • Figure 14 further depicts the correlation between FRα plasma concentrations determined using EIA and ECLIA. The correlation is high (r=0.95).
  • EXAMPLE 13. DETERMINATION OF FRα LEVELS IN MATCHED URINE AND SERUM SAMPLES FROM LUNG CANCER PATIENTS AND OVARIAN CANCER PATIENTS AS MEASURED BY ELECTROCHEMILUMINESCENCE IMMUNOASSAY (ECLIA)
  • FRα levels were determined in matched urine and serum samples from lung cancer and ovarian cancer patients using ECLIA where the samples were taken from the same patient. The correlation between serum and urine FRα levels was also determined.
  • Materials and Methods
  • The ECLIA methodology utilized is as described in Example 1. Guanidine was used to solubilize urine sediments as described in Example 6.
  • Results
  • The results of the ECLIA assays of serum and urine from lung cancer and ovarian cancer patients are presented in Table 22. Table 22: FRα concentrations in matched urine and serum samples of lung cancer patients and ovarian cancer patients, as determined by ECLIA
    Serum Urine
    Group set ID pg/mL pg/mL
    Lung cancer
    1 146 2009
    2 153 4496
    3 206 -
    4 70 3562
    5 195 12381
    6 352 21873
    7 198 11296
    8 120 18570
    9 275 4455
    10 163 8662
    11 145 5294
    12 178 -
    13 165 1106
    14 187 7446
    15 168 11167
    16 217 24448
    17 142 6724
    18 177 14514
    19 236 822
    20 101 4826
    21 145 7723
    22 213 9887
    23 143 7422
    24 253 3376
    25 421 8045
    ovarian cancer 26 282 9414
    27 1605 7651
    28 240 13059
    29 695 10549
  • Summary data for serum and urine FRα levels of lung cancer patients is presented in Table 23. Table 23: Summary statistics for FRα concentrations in matched serum and urine samples of lung cancer patients, as determined by ECLIA
    Lung cancer FRα (pg/mL)
    Serum Urine
    n 25 23
    Mean 191 8700
    SD 75 6291
    Max. 421 24448
    Min. 70 822
  • Summary data for serum and urine FRα levels of ovarian cancer patients is presented in Table 24. Table 24: Summary statistics for FRα concentrations in matched serum and urine samples of ovarian cancer patients, as determined by ECLIA
    Ovarian cancer FRα (pg/mL)
    Serum Urine
    n
    4 4
    Mean 705 10168
    SD 634 2266
    Max. 1605 13059
    Min. 240 7651
  • Figure 15 shows correlations between ECLIA measures of FRα levels in matched serum and urine samples taken from the same patient. The correlation for lung cancer patients was r=0.24 (upper panel) and the correlation for ovarian cancer patients was r=-0.76 (lower panel).
  • These data demonstrate the relative lack of correlation between FRα concentrations measured in urine versus serum, especially as shown for lung cancer patients. Further, these data demonstrate that FRα is basically non-detectable above background levels in the serum of lung cancer patients versus normal controls whereas FRα is detectable in the urine of these patients.
  • EXAMPLE 14. ASSESSMENT OF LEVELS OF FRα IN SERUM SAMPLES FROM PATIENTS WITH OVARIAN CANCER, PATIENTS WITH LUNG CANCER, AND NORMAL CONTROLS
  • FRα levels in the serum of patients with ovarian cancer, patients with lung cancer, and normal controls were assessed. Serum FRα levels were assessed using ECLIA with two different pairs of capture-detector antibodies: Pair 1, in which 9F3 was the capture antibody and 24F12 was the detector antibody, and Pair 2, in which 26B3 was the capture antibody and 19D4 was the detector antibody.
  • The FRα pairs were tested with full calibrator curves and 196 individual serums diluted 1:4. In one experiment, 26B3 was used as the capture antibody after CR processing on a plate lot (75 µg/mL, +B, +T) and 19D4 was used as the detector antibody at 1.0µg/mL. In another experiment, 9F3 was used as the capture antibody and 24F12 was used as the detector antibody at 1.0µg/mL. Each were CR processed (lot 10070) with an label to protein ratio (L/P) of 13.3. Diluent 100 (Meso Scale Discovery, Gaithersburg, Maryland) + human anti-mouse antibody (HAMA) +mIgG was used for samples and calibrator. Diluent 3 (Meso Scale Discovery, Gaithersburg, Maryland) was used for detections.
  • The following protocol was employed for the ECLIA. Samples were added at 50µL/well. The samples were shaken for 2 hours and subsequently washed with Phosphate Buffered Saline (PBS) solution with the detergent Tween 20 (PBST). The detector antibody was added at 25µL/well. The samples were shaken for 2 hours and then were washed with PBST. Finally, the electrochemiluminescence (ECL) emission of the samples was read with 2X MSD® Buffer T.
  • The results are shown in Table 25 below. Table 25: FRα levels in serum of patients with ovarian cancer, patients with lung cancer, and normal controls
    LLOQ1 = 1pg/m L LLOQ1 = 5 pg/mL
    FRα- Pair 2 FRα Pair 1
    MSD Sample Testing Number Sample Type Adjusted backfit conc2 (pg/mL) %CV Adjusted backfit conc2 (pg/mL) %CV stage grade gender comments
    1 Ovarian Serum 3760 4% 3585 4% III 2 F Adenocarcinoma-Ovary
    2 Ovarian Serum 223 3% 273 2% III 3 F Aderiocarcinoma-Ovary
    3 Ovarian Serum 950 1% 3346 8% IIIC F Papillary Serous Carcinoma
    4 Ovarian Serum 3827 4% 968 0% III 2 F Adenocarcinoma-Ovary
    5 Ovarian Serum 251 6% 468 2% IV 2 F Aderiocarcinoma-Ovary
    6 Ovarian Serum 199 6% 328 1% IIIC 2 F Cystaderiocarcinoma
    7 Ovarian Serum 166 1% 257 5% IC 2 F Cystaderiocarcinoma
    8 Ovarian Serum 182 4% 248 2% IIIC 2 F Cystaderiocarcinoma
    9 Ovarian Serum 155 6% 265 2% IIIC 2 F Cystaderiocarcinoma
    10 Ovarian Serum 145 9% 253 5% IIIC 2 F Cystaderiocarcinoma
    11 Ovarian Serum 142 5% 186 1% IIB 1 F Serous adenocarcinoma
    12 Ovarian Serum 299 5% 456 2% IB 3 F Serous adenocarcinoma
    13 Ovarian Serum 315 0% 768 8% IIIB high grade F Serous cystaderiocarcinoma of the ovary
    14 Ovarian Serum 168 6% 351 1% I high grade F Serous cystaderiocarcinoma of the ovary
    15 Ovarian Serum 187 8% 263 1% I Well to moderately differentiated F Papillary serous cystaderiocarcinoma of the ovary
    16 Ovarian Serum 423 3% 229 5% IA poorly differentiated F Mucinous cystadenocarcinoma of the ovary
    17 Ovarian Serum 86 7% 289 1% I moderately differentiated F Papillary cystadenocarcinoma of the ovary
    18 Ovarian Serum 57 3% 255 1% I well differentiated F Mucinous cystaderiocarcinoma of the ovary
    19 Ovarian Serum 108 1% 393 1% III moderately differentiated F Papillary mucinous cystaderiocarcinoma of the ovary
    20 Ovarian Serum 207 3% 328 1% missing poorly differentiated F Papillary cystaderiocarcinoma of the ovary
    21 Ovarian Serum 66 0% 254 1% missing high grade F Aderiocarcinoma of the ovary
    22 Ovarian Serum 108 2% 197 2% III low grade F Papillary cystadenocarcinoma of the ovary
    23 Ovarian Serum 201 2% 457 1% II high grade F Papillary serous cystaderiocarcinoma of the ovary
    24 Ovarian Serum 477 1% 808 11% III high grade F Serous cystaderiocarcinoma of the ovary
    25 Ovarian Serum 423 3% 613 3% n/a n/a F transitional cell carcinoma
    26 Ovarian Serum 249 8% 432 1% n/a 2 F ovarian carcinoma - endometrioid type
    27 Ovarian Serum 152 7% 235 2% IV 2 F ovarian carcinoma - serous papillary type
    28 Ovarian Serum 1409 5% 1287 3% III C 3 F ovarian carcinoma - serous type
    29 Ovarian Serum 443 2% 547 2% III C 2 F ovarian carcinoma - serous papillary type
    30 Ovarian Serum 208 1% 298 13% IA 1 F ovarian carcinoma - serous type
    31 Ovarian Serum 114 9% 344 1% III B 2 F ovarian carcinoma - serous papillary type
    32 Ovarian Serum 223 4% 157 133% III C 1 F ovarian carcinoma - serous papillary type
    33 Ovarian Serum 5034 1% 4405 1% III C 3 F transitional cell carcinoma
    34 Ovarian Serum 32966 6% 23228 4% IV n/a F ovarian carcinoma - serous papillary type
    35 Ovarian Serum 94 6% 188 3% n/a 3 F clear cell adenocarcinoma
    36 Ovarian Serum 866 6% 1317 1% III C 3 F poorly differentiated adenocarcinoma
    37 Ovarian Serum 2916 8% 3121 0% IV 3 F ovarian carcinoma - serous papillary type
    38 Ovarian Serum 679 4% 1037 17% IV 3 F ovarian carcinoma - serous type
    39 Ovarian Serum 294 8% 478 3% III C 3 F ovarian carcinoma - serous papillary type
    40 Ovarian Serum 2037 4% 12 74% III C 3 F ovarian carcinoma - serous type
    41 Ovarian Serum 16289 6% 10431 3% IIIC 2 F ovarian carcinoma - serous papillary type
    42 Ovarian Serum 386 6% 736 6% n/a 3 F ovarian carcinoma - serous papillary type
    43 Ovarian Serum 1474 6% 2382 3% IIIC F Papillary serous cystadenocarcinoma of the pelvic
    44 Ovarian Serum 169 2% 438 1% I 3 F Aderiocarcinoma of the ovary
    45 Ovarian Serum 257 4% 635 10% I high grade F Adenocarcinoma of the ovary
    46 Ovarian Serum 184 6% 393 0% IIIC poorly differentiated F Aderiocarcinoma of the ovary
    47 Ovarian Serum 251 6% 659 1% IIA 2 F Aderiocarcinoma of the ovary
    48 Ovarian Serum 165 2% 394 7% I 2 F Adenocarcinoma of the ovary
    49 Ovarian Serum 64 7% 245 6% IIIB undifferentiated F Serous cystadenocarcinoma of the ovary
    50 Ovarian Serum 90 3% 203 3% IIIC F Serous adenocarcinoma nos
    51 Ovarian Serum 167 3% 376 11% IIIC high grade F Serous papillary adenocarcinoma nos
    52 Ovarian Serum 112 4% 260 5% IIIC high grade F Serous adenocarcinoma nos
    53 Ovarian Serum 198 2% 340 6% IV high grade F Serous cystadenocarcinoma, nos
    54 Ovarian Serum 134 3% 363 6% I moderately differentiated F Papillary mucinous cystaderiocarcinoma of the ovary
    55 Ovarian Serum 118 5% 304 10% IB high grade F Papillary serous cystaderiocarcinoma of the ovary
    56 Ovarian Serum 107 1% 326 4% IIIB high grade F Papillary serous cystadenocarcinoma of the ovary
    57 Ovarian Serum 728 1% 1670 2% IIIB F Papillary serous cystaderiocarcinoma of the ovary
    58 Ovarian Serum 2138 1% 3257 2% IIIC F Papillary adenocarcinoma of the ovary
    59 Ovarian Serum 167 4% 410 1% III F Papillary serous cystaderiocarcinoma of the ovary
    60 Ovarian Serum 3054 4% 3285 14% IIIC high grade F Serous carcinoma of the ovary
    61 Ovarian Serum 97 3% 215 0% IIIB high grade F Papillary serous cystaderiocarcinoma of the ovary
    63 Lung Serum 61 3% 366 10% IA moderate to poorly differentiated M Aderiocarcinoma of the lung
    64 Lung Serum 106 1% 319 4% IB M Aderiocarcinoma of the lung
    65 Lung Serum 49 2% 257 1% IB moderately differentiated M Adenocarcinoma of the lung
    66 Lung 75 2% 301 2% II 3 M Aderiocarcinoma
    Serum
    67 Lung Serum 79 3% 283 8% II 2 M Adenocarcinoma
    68 Lung Serum 145 2% 492 0% II 2 F Aderiocarcinoma
    69 Lung Serum 142 2% 367 6% IV n/a F adenocarcinoma
    70 Lung Serum 103 2% 231 7% IV n/a F adenocarcinoma
    71 Lung Serum 153 1% 311 3% III B n/a F adenocarcinoma
    72 Lung Serum 54 3% 124 6% III A missing M large and solid cell carcinoma
    73 Lung Serum 183 3% 391 10% III B missing F adenocarcinoma
    74 Lung Serum 91 2% 199 7% missing 3 M poorly differentiated non-keratinizing squamous cell carcinoma
    75 Lung Serum 89 7% 221 2% III B 3 F poorly differentiated adenocarcinoma
    76 Lung Serum 139 1% 330 1% IA 2 F moderately differentiated adenocarcinoma
    77 Lung Serum 197 3% 452 7% III A 2 F moderately differentiated adenocarcinoma
    78 Lung Serum 52 3% 183 5% III A n/a F pleomorphic carcinoma
    79 Lung Serum 80 3% 249 8% III A n/a M pleomorphic carcinoma
    80 Lung Serum 72 1% 158 7% IIIA 2 F moderately differentiated adenocarcinoma
    81 Lung Serum 130 12% 221 1% IA 3 M large and solid cell carcinoma
    82 Lung Serum 81 6% 155 1% IB missing M large and solid cell carcinoma
    83 Lung Serum 127 4% 278 3% IIIB missing F large and solid cell carcinoma
    84 Lung Serum 129 3% 240 2% missing missing F large and solid cell carcinoma
    85 Lung Serum 135 2% 231 7% III B 2 M moderately differentiated adenocarcinoma
    86 Lung Serum 235 1% 330 0% IV 2 M adenocarcinoma
    87 Lung Serum 243 5% 396 2% IV 3 F poorly differentiated adenocarcinoma
    88 Lung Serum 204 3% 572 6% IA moderately differentiated F Squamous cell carcinoma of the lung
    89 Lung Serum 54 9% 214 4% IB moderately differentiated M Aderiocarcinoma of the lung
    90 Lung Serum 116 7% 270 0% IB moderately differentiated F Mucinous adenocarcinoma of the lung
    91 Lung Serum 117 3% 292 5% IIA moderately differentiated M Adenocarcinoma of the lung
    92 Lung Serum 248 1% 578 2% missing moderately differentiated M Aderiocarcinoma of the lung
    93 Lung Serum 86 2% 300 9% IB poorly differentiated M Adenocarcinoma of the lung
    94 Lung Serum 33 6% 117 5% IIIA moderate to poorly differentiated M Aderiocarcinoma of the lung
    95 Lung Serum 36 3% 196 3% IIIA poorly differentiated M Aderiocarcinoma of the lung
    96 Lung Serum 237 3% 722 4% IB well differentiated M Adenocarcinoma of the lung
    97 Lung Serum 82 9% 286 2% IB poorly differentiated M Adenocarcinoma of the lung
    98 Lung Serum 112 1% 431 0% IB Well to moderately differentiated F Aderiocarcinoma of the lung
    99 Lung Serum 137 5% 379 0% IA moderately differentiated F Alveolar adenocarcinoma of the lung
    100 Lung Serum 65 2% 181 9% IA moderate to poorly differentiated M Aderiocarcinoma of the lung
    101 Lung Serum 119 6% 280 4% IB M Aderiocarcinoma of the lung
    102 Normal Serum 187 6% 391 2% F
    103 Normal Serum 337 4% 560 6% F
    104 Normal Serum 203 4% 405 3% F
    105 Normal Serum 97 3% 311 2% F
    106 Normal Serum 210 11% 393 6% F
    107 Normal Serum 135 9% 271 6% M
    108 Normal Serum 145 2% 225 3% F
    109 Normal Serum 182 4% 257 5% M
    110 Normal Serum 186 5% 297 2% M
    111 Normal Serum 129 8% 197 4% M
    112 Normal Serum 133 4% 254 4% M
    113 Normal Serum 136 1% 298 9% M
    114 Normal Serum 189 3% 321 1% M
    115 Normal Serum 167 1% 257 6% M
    116 Normal Serum 159 1% 315 1% M
    117 Normal Serum 166 3% 270 1% M
    118 Normal Serum 197 2% 339 0% M
    119 Normal Serum 148 1% 389 4% M
    120 Normal Serum 198 4% 833 9% M
    121 Normal Serum 101 3% 137 0% M
    122 Normal Serum 111 8% 266 1% M
    123 Normal Serum 96 0% 172 2% M
    124 Normal Serum 224 1% 249 7% M
    125 Normal Serum 203 8% 312 1% M
    126 Normal Serum 277 10% 388 5% M
    127 Normal Serum 191 4% 272 4% F
    128 Normal Serum 206 4% 297 3% F
    129 Normal Serum 204 15% 194 3% F
    130 Normal Serum 156 1% 106 2% F
    131 Normal Serum 177 3% 195 3% F
    132 Normal Serum 109 0% 148 5% M
    134 Normal Serum 116 1% 281 1% F
    135 Normal Serum 182 7% 250 6% M
    136 Normal Serum 324 2% 475 7% F
    137 Normal Serum 122 18% 191 1% F
    138 Normal Serum 135 7% 185 1% F
    139 Normal Serum 264 4% 372 2% F
    140 Normal Serum 105 4% 188 5% M
    141 Normal Serum 374 0% 649 0% M
    142 Normal Serum 93 7% 162 7% M
    143 Normal Serum 143 7% 326 4% F
    144 Normal Serum 108 3% 202 2% F
    145 Normal Serum 153 8% 341 3% F
    146 Normal Serum 448 8% 400 5% F
    147 Normal Serum 109 4% 196 3% F
    148 Normal Serum 142 6% 218 1% F
    149 Normal Serum 174 4% 309 9% F
    150 Normal Serum 185 5% 270 2% F
    151 Normal Serum 180 3% 241 0% M
    152 Normal Serum 125 5% 314 4% F
    153 Normal Serum 270 0% 449 2% F
    154 Normal Serum 127 9% 232 1% M
    155 Normal Serum 251 3% 415 6% F
    156 Normal Serum 121 1% 349 0% M
    157 Normal Serum 137 8% 223 0% M
    158 Normal Serum 77 3% 173 6% M
    159 Normal Serum 143 4% 223 7% F
    160 Normal Serum 121 5% 411 8% M
    161 Normal Serum 99 8% 199 3% F
    162 Normal Serum 158 2% 236 0% F
    163 Normal Serum 138 7% 235 3% F
    164 Normal Serum 175 18% 290 2% F
    165 Normal Serum 339 4% 589 8% M
    166 Normal Serum 155 4% 372 1% F
    167 Normal Serum 166 0% 278 1% M
    168 Normal Serum 231 7% 377 3% M
    169 Normal Serum 148 10% 255 3% F
    170 Normal Serum 172 2% 312 4% M
    171 Normal Serum 146 6% 344 1% M
    172 Normal Serum 158 3% 306 4% M
    173 Normal Serum 145 2% 274 6% F
    174 Normal Serum 163 12% 279 1% M
    175 Normal Serum 83 5% 196 0% M
    176 Normal Serum 102 7% 282 5% M
    177 Normal Serum 140 3% 330 6% M
    178 Normal Serum 174 9% 277 15% F
    179 Normal Serum 295 3% 281 8% M
    180 Normal Serum 67 4% 308 13% F
    181 Normal Serum 115 3% 324 0% F
    182 Normal Serum 128 5% 287 0% F
    183 Normal Serum 128 1% 112 79% M
    184 Normal Serum 76 3% 147 53% F
    185 Normal Serum 264 7% 377 4% F
    186 Normal Serum 146 13% 258 3% M
    187 Normal Serum 132 0% 264 1% F
    188 Normal Serum 92 4% 249 1% M
    189 Normal Serum 89 11% 251 4% F
    190 Normal Serum 135 5% 268 4% M
    191 Normal Serum 177 4% 394 3% F
    192 Normal Serum 184 8% 367 2% F
    193 Normal Serum 156 3% 387 5% F
    194 Normal Serum 118 8% 275 4% M
    195 Normal Serum 74 7% 217 6% M
    196 Normal Serum 185 8% 373 5% F
    197 Normal Serum 159 3% 378 2% F
    198 Normal Serum 94 2% 245 1% F
    1 LLOQ is the lower limit of quantitation
    2 The adjusted backfit concentration is adjusted to take into account the sample dilution.
  • Based on the foregoing data, it was apparent that the 9F3, 2412, 26B3 and 19D4 antibodies were useful in detecting levels of FRα in biological samples, for example serum, derived from a subject. Moreover, the particular combinations of (i) 9F3 as a capture antibody and 24F12 as a detector antibody and (ii) 26B3 as a capture antibody and 19D4 as a detector antibody were capable and particularly effective of assessing levels of FRα in biological samples.
  • EXAMPLE 15. ASSESSMENT OF LEVELS OF FRα IN URINE USING THREE DIFFERENT DETECTOR AND CAPTURE ANTIBODY PAIRS
  • The ability of three anti-FRα antibody pairs in detecting the levels of FRα in urine samples was assessed. The antibody pairs utilized were as follows: (1) 26B3 as detector antibody and 9F3 as capture antibody, and (2) 24F12 as detector antibody and 9F3 as capture antibody.
  • Method
  • Two antibody pairs were tested with full calibrator curves and urine pretreated with a 1:1 dilution for 2 minutes in either 6M guanidine, 3M guanidine or PBS control. The following urine samples were tested: three human urine pools diluted 1:80, and five human individual urines diluted 1:80 (one male, four female).
  • Plates were Biodotted at 150µg/mL, +B, +T, on 4spot STD ((Meso Scale Discovery, Gaithersburg, Maryland)), one capture per well. Detects were run at 1 µg/mL. Diluent 100 + HAMA + mIgG was used for samples and calibrator. Diluent 3 was used for detections. Diluents were commercially available diluents obtained from Meso Scale Discovery.
  • The following protocol was employed for the ECLIA. Samples were added at 50µL/well. The samples were shaken for 2 hours. The samples were washed with Phosphate Buffered Saline (PBS) solution with the detergent Tween 20 (PBST). The detector antibody was added at 25µL/well. The samples were shaken for 2 hours and subsequently washed with PBST. The electrochemiluminescence (ECL) emission of the samples was read with 2X MSD Buffer T.
  • The results of these experiments are shown in Tables 26-27. Table 26: Detection of FRα levels in urine using 26B3 as detector antibody and 9F3 as capture antibody
    Detect 26B3
    Capture 9F3
    6M Guanidine 3M Guanidine PBS Control
    Sample ID Adjusted Backfit conc pg/mL %CV % of control Adjusted Backfit conc pq/mL %CV % of control Adjusted Backfit conc pg/mL %CV
    Urine pool 1 13,252 2% 114% 14,505 10% 124% 11,655 17%
    Urine pool 2 14,827 5% 133% 17,039 4% 152% 11,187 5%
    Urine pool 3 11,280 5% 119% 9,065 10% 96% 9,479 9%
    Urine Ind 1 1,747 3% 99% 1,754 6% 100% 1,760 12%
    Urine Ind 2 40,505 7% 145% 46,622 5% 167% 27,920 13%
    Urine Ind 3 1,623 1% 117% 1,496 5% 108% 1,381 4%
    Urine Ind 4 12,091 2% 86% 14,941 5% 107% 13,996 13%
    Urine Ind 5 22,829 2% 128% 24,607 8% 137% 17,899 5%
    Average 3% 118% Average 7% 124% Average 10%
    difference difference difference from 6M condition 6%
    Table 27: Detection of FRα levels in urine using 24F12 as detector antibody and 9F3 as capture antibody
    Detect 24F12
    Capture 9F3
    6M Guanidine 3M Guanidine PBS Control
    Sample ID Adjusted Backfit conc pg/mL %CV % of control Adjusted Backfit conc pg/mL %CV % of control Adjusted Backfit conc pg/mL %CV
    Urine pool 1 10,883 2% 53% 14,689 9% 72% 20,504 10%
    Urine pool 2 11,763 8% 60% 16,487 7% 85% 19,468 1%
    Urine pool 3 7,456 9% 40% 9,362 17% 50% 18,677 7%
    Urine Ind 1 1,376 1% 39% 1,894 7% 54% 3,501 3%
    Urine Ind 2 29,567 0% 61% 37,843 13% 78% 48,607 5%
    Urine Ind 3 1,621 4% 61% 2,153 2% 81% 2,667 1%
    Urine Ind 4 10,470 6% 58% 14,116 5% 78% 18,175 6%
    Urine Ind 5 19,390 6% 68% 22,076 23% 78% 28,421 4%
    Average 5% 55% Average 10% 72% Average 5%
    difference difference difference from 6M condition 17%
  • Based on the foregoing data, it was apparent that the 9F3, 24F12, 26B3 and 19D4 antibodies were useful in detecting levels of FRα in biological samples derived from a subject. Moreover, the combinations of (1) 26B3 as detector antibody and 9F3 as capture antibody and (2) 24F12 as detector antibody and 9F3 as capture antibody were capable and particularly effective of assessing levels of FRα in biological samples.
  • A second set of experiments, following the protocol described above and using the same two pairs of antibodies, were conducted utilizing four female human individual urines diluted 1:80. The urine was pretreated with a 1:1 dilution for 2 minutes in either 3M guanidine or PBS control. The results are shown in Tables 28-29. Table 28: Detection of FRα levels in urine using 26B3 as detector antibody and 9F3 as capture antibody
    Detect 26B3
    Capture 9F3
    3M Guanidine PBS Control
    Sample ID Adjusted Backfit conc pg/mL %CV % of control Adjusted Backfit conc pg/mL %CV
    Urine Ind
    2 33,824 4% 98% 34,569 2%
    Urine Ind
    3 2,086 4% 99% 2,107 3%
    Urine Ind
    4 15,283 5% 97% 15,696 2%
    Urine Ind
    5 24,955 4% 92% 26,991 3%
    Average 4% 97% Average 3%
    Table 29: Detection of FRα levels in urine using 24F12 as detector antibody and 9F3 as capture antibody
    Detect 24F12
    Capture 9F3
    3M Guanidine PBS Control
    Sample ID Adjusted Backfit conc pg/mL %CV % of control Adjusted Backfit conc pg/mL %CV
    Urine Ind
    2 38,455 4% 106% 36,414 6%
    Urine Ind
    3 2,447 2% 109% 2,250 2%
    Urine Ind
    4 15,303 3% 81% 18,964 8%
    Urine Ind
    5 27,216 0% 95% 28,651 5%
    Average 2% 98% Average 5%
  • The results of this second set of experiments further confirm the results of the first set of experiments and demonstrate that the level of FRα which is not bound to a cell can be reliably assessed, for example, in urine, using assays such as the ECLIA assay and using the 26B3, 9F3, 24F12 antibodies. Further, the results demonstrate that such assays can effectively detect FRα using pairs of detector and capture antibodies that bind FRα (such as, e.g., 26B3 as detector antibody and 9F3 as capture antibody, and 24F12 as detector antibody and 9F3 as capture antibody).
  • EXAMPLE 16. ASSESSMENT OF LEVELS OF FRα IN SERUM AND PLASMA
  • The levels of FRα were assessed in samples of serum and plasma on two separate days. The subjects from whom the samples were derived were either normal subjects or patients with ovarian or lung cancer.
  • The protocol for assessing FRα levels was the same as set forth in Example 14 above. The pairs of antibodies used for assessing FRα levels were also the same as in Example 14, i.e., Pair 1, in which 9F3 was the capture antibody and 24F12 was the detector antibody, and Pair 2, in which 26B3 was the capture antibody and 19D4 was the detector antibody.
  • The results are provided in Table 30. Table 30: Levels of FRα as assessed in serum and plasma samples on different days
    Day 1 Day 2 Day 1 Day 2 Day 1 Day 2 Day 1 Day 2
    FRα/pair1 FRα/pair1 FRα/pair1 FRα/pair1 FRα/pair2 FRα/pair2 FRα/pair2 FRα/pair2
    Donor ID Disease Biosample Confirmed Diagnosis Stage Serum (pg/ml) Serum (pg/ml) Plasma (pg/ml) Plasma (pg/ml) Serum (pg/ml) Serum (pg/ml) Plasma (pg/mL) Plasma (pg/mL)
    17168 ovary Serum FRA - Pair 1 versus Pair 2 I 1236 1282 1466 1183 1298 1384 1410 1336
    46464 ovary Serous carcinoma IIIC 1589 1848 2027 2147 1966 2018 2066 2210
    47219 ovary Adenocarcinoma missing 447 432 1208 748 435 446 695 577
    47721 ovary Papillary serous cystadenocarcinoma IIIB 1307 1400 2291 2100 1642 1479 1940 1807
    48185 ovary Adenocarcinoma missing 1058 1038 883 652 918 872 811 781
    48254 ovary Adenocarcinoma IIIC 511 495 3030 2569 506 445 1332 1370
    48258 ovary Adenocarcinoma missing 471 552 1978 1070 629 547 978 798
    48282 ovary Adenocarcinoma missing 375 388 688 536 446 407 540 428
    48698 ovary Carcinoma, undifferentiated IIIB 279 231 308 225 340 255 328 228
    49028 ovary Serous carcinoma IIIC 727 590 158 169 468 526 192 192
    49030 ovary Serous carcinoma IIIC 215 205 695 485 362 291 734 590
    49033 ovary Serous cystadenocarcinoma IIIC 579 457 805 717 511 451 629 668
    49071 ovary Serous cystadenocarcinoma IV 335 338 1884 1462 559 457 1224 1028
    49092 ovary Papillary cystadenocarcinoma missing 272 228 1235 615 399 334 678 535
    49258 ovary Papillary serous cystadenocarcinoma IIIB 201 190 343 142 328 253 368 244
    49335 ovary Serous cystadenocarcinoma IIIB 1904 1698 1560 1485 1879 1503 1868 1627
    49369 ovary Serous carcinoma IIIC 2451 2805 2589 2641 3402 2659 2898 2605
    49551 ovary Serous carcinoma III 408 385 364 342 505 432 436 418
    50009 ovary Serous carcinoma IIIC 2887 4127 2641 3811 4466 3914 3533 3519
    50370 ovary Serous cystadenocarcinoma IIIB 619 570 788 611 813 762 708 758
    50378 ovary Papillary serous cystadenocarcinoma I 410 387 330 256 388 464 381 387
    50460 ovary Papillary serous cystadenocarcinoma IIIB 254 274 688 414 284 272 489 420
    50467 ovary Clear cell adenocarcinoma I 427 314 414 300 272 313 292 309
    50635 ovary Serous cystadenocarcinoma IA 247 227 462 367 355 345 392 392
    51503 ovary Papillary cystadenocarcinoma I 291 250 925 651 315 326 818 498
    51504 ovary Mucinous cystadenoma, borderline malignancy I 224 184 2438 1395 225 240 916 856
    51506 ovary Papillary mucinous cystadenocarcinoma III 270 258 713 324 335 384 457 424
    52949 ovary Adenocarcinoma missing 446 436 288 257 435 483 395 390
    52952 ovary Papillary serous cystadenocarcinoma missing 480 415 451 376 310 351 299 321
    52957 ovary Papillary serous cystadenocarcinoma III 373 334 274 221 318 374 303 330
    52978 ovary Papillary mucinous cystadenocarcinoma II 348 318 533 429 525 559 596 624
    52980 ovary Serous cystadenocarcinoma III 627 630 1447 1019 834 905 964 996
    DLSN-057 normal 309 402 413 394 515 527 509 531
    DLSN-056 normal 273 254 483 498 480 458 522 524
    DLSN-052 normal 282 289 293 298 446 483 438 401
    DLSN-049 normal 282 256 351 320 363 394 430 359
    DLSN-048 normal 362 399 454 434 746 722 824 639
    DLSN-047 normal 188 176 208 167 275 263 251 212
    DLSN-046 normal 295 321 276 240 354 310 315 257
    DLSN-045 normal 259 259 259 189 292 279 273 235
    DLSN-044 normal 244 236 246 254 284 273 284 271
    DLSN-042 normal 245 199 210 185 328 301 285 258
    DLSN-040 normal 392 376 408 406 481 499 502 413
    DLSN-039 normal 463 470 469 446 617 599 567 528
    DLSN-037 normal 256 231 256 223 367 338 351 289
    DLSN-029 normal 285 270 348 337 453 417 418 351
    DLSN-023 normal 265 254 286 298 458 402 370 386
    DLSN-020 normal 237 238 311 287 530 496 504 446
    DLSN-011 normal 344 328 210 207 688 603 297 267
    DLSN-039 normal 196 178 204 183 259 219 242 248
    DLSN-047 normal 336 385 489 429
    DLSN-052 normal 226 208 346 315
    DLSL-012 lung 340 220 529 375
    DLSL-015 lung 199 180 236 292
    DLSL-023 lung 581 347 568 523
    DLSL-031 lung 400 251 415 343
    DLSL-034 lung 133 138 299 331
    1 lung 238 339 462 460
    3 lung 120 185 203 334
    6 lung 295 1646 431 868
    18 lung 328 288 466 418
    18639 lung 124 333 128 216
    18640 lung 246 275 221 221
    50666 lung 372 357 405 338
    2 lung 214 327 274 269
    4 lung 347 625 526 575
    7 lung 224 308 421 452
    12 lung 409 424 778 725
    13 lung 509 590 1251 1360
    14 lung 250 250 409 357
    15 lung 502 466 606 594
    16 lung 153 274 246 275
    19 lung 357 905 428 576
    DLS4-0002 lung 234 245 410 347
    DLS4-0004 lung 472 520 507 538
    DLSO-007 ovary 389 593 574 544
    DLSO-008 ovary 175 295 386 364
    DLSO-009 ovary 232 331 299 349
    DLSO-014 ovary 265 300 430 467
    DLSO-020 ovary 536 261 453 389
    DLSO-026 ovary
    DLSO-027 ovary 290 140 269 218
    DLSO-028 ovary 357 207 397 18
    DLSO-029 ovary 412 236 369 361
    DLSO-030 ovary 387 498 595 616
    DLSO-031 ovary 348 496 353 413
    DLSO-034 ovary 197 222 308 346
    DLSO-035 ovary 144 540 298 587
    DLSO-023 ovary 404 216 423 337
    DLSO-025 ovary 306 156 301 272
    DLSO-026 ovary 231 293 406 378
    DLSO-032 ovary 151 190 240 255
    DLSO-018 ovary 155 340 303 343
    DLSO-019 ovary 350 330
    DLSO-021 ovary 359 365 346 434
    DLSO-024 ovary 260 140 338 318
    10001627 ovary 1481 1396 1583 1655
    11025393 ovary 5241 4069 4998 5486
    11025394 ovary 473 371 488 518
    110025395 ovary 3215 2920 3466 4043
    110025397 ovary 109 145 245 232
    110025398 ovary 476 497 613 543
    110025399 ovary 166 145 234 229
    110025402 ovary 219 221 389 357
    110025403 ovary 7529 6990 13033 11888
    110025405 ovary 510 508 836 835
    5 ovary 164 328 252 375
    8 ovary 251 709 473 573
    9 ovary 249 743 482 561
    10 ovary 288 569 412 446
    11 ovary 180 417 310 397
    17 ovary 296 760 342 628
    110025392 ovary 1094 2210 907 1294
  • Based on the foregoing data, it was apparent that the 9F3, 2412, 26B3 and 19D4 antibodies were useful in detecting levels of FRα in biological samples, for example, serum or plasma, derived from a subject. Moreover, the particular combinations of (i) 9F3 as a capture antibody and 24F12 as a detector antibody and (ii) 26B3 as a capture antibody and 19D4 as a detector antibody were capable and particularly effective in assessing levels of FRα in biological samples.
  • For assays conducted using both Pair 1 and Pair 2, there was a high correlation between serum and plasma FRα levels. Figure 16 shows the correlation in serum versus plasma FRα levels for assays conducted using Pair 1 (see Example 16). The R2 value was 0.8604. Figure 17 shows the correlation in serum versus plasma FRα levels for assays conducted using Pair 2 (see Example 16). The R2 value was 0.9766.
  • For both serum and plasma samples, there was a high correlation between FRα levels measured using Pair 1 and Pair 2. Figure 18 shows the correlation in serum FRα levels for assays conducted using Pair 1 versus Pair 2 (see Example 16). The R2 value was 0.9028. Figure 19 shows the correlation in plasma FRα levels for assays conducted using pair 1 versus pair 2 (see Example 16). The R2 value was 0.8773.
  • The results also showed that there was a high correlation between FRα levels measured on different days. Figure 20 shows the interday correlation in serum FRα levels for assays conducted using pair 2. The R2 value was 0.9839.
  • EQUIVALENTS
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims. Any combination of the embodiments disclosed in the dependent claims are contemplated to be within the scope of the invention. Table 33: SEQUENCES
    SEQ ID NO: DESCRIPTION SEQUENCE
    1 MORAB-003 CDRH1 GFTFSGYGLS
    2 MORAB-003 CDRH2 MISSGGSYTYYADSVKG
    3 MORAB-003 CDRH3 HGDDPAWFAY
    4 MORAB-003 CDRL1 SVSSSISSNNLH
    5 MORAB-003 CDRL2 GTSNLAS
    6 MORAB-003 CDRL3 QQWSSYPYMYT
    7 MORAb-003 Heavy Chain Mature Polypeptide Amino Acid Sequence
    Figure imgb0005
    8 MORAb-003 Light Chain Mature Polypeptide Amino Acid Sequence
    Figure imgb0006
    9 MORAb-003 Heavy Chain full length pre-protein amino acid sequence
    Figure imgb0007
    10 MORAb-003 Light Chain full length pre-protein amino acid sequence
    Figure imgb0008
    11 MORAb-003 Heavy Chain Nucleotide
    Figure imgb0009
    Figure imgb0010
    12 MORAb-003 Light Chain Nucleotide
    Figure imgb0011
    13 LK26HuVK
    Figure imgb0012
    14 LK26HuVKY
    Figure imgb0013
    15 LK26HuVKPW
    Figure imgb0014
    16 LK26HuVKPW, Y
    Figure imgb0015
    17 LK26HuVH
    Figure imgb0016
    Figure imgb0017
    18 LK26HuVHFAIS, N
    Figure imgb0018
    19 LK26HuVHSLF
    Figure imgb0019
    20 LK26HuVHI,
    Figure imgb0020
    21 LK26KOLHuVH
    Figure imgb0021
    22 Murine LK26 Ab Heavy Chain Sequence
    Figure imgb0022
    23 Murine LK26 Ab Light Chain Sequence
    Figure imgb0023
    24 Consensus Human FRα Nucleotide Sequence
    Figure imgb0024
    25 Consensus Human FRα Amino Acid Sequence
    Figure imgb0025
    26 Mov-18 TELLNVXMNAK*XKEKPXPX
    epitope sequence *KLXXQX
    27 9F3 Light Chain CDR1 RASSTVSYSYLH
    28 9F3 Light Chain CDR2 GTSNLAS
    29 9F3 Light Chain CDR3 QQYSGYPLT
    30 9F3 Light Chain Variable Domain
    Figure imgb0026
    31 9F3 Heavy Chain CDR1 SGYYWN
    32 9F3 Heavy Chain CDR2 YIKSDGSNNYNPSLKN
    33 9F3 Heavy Chain CDR3 EWKAMDY
    34 9F3 Heavy Chain Variable Domain
    Figure imgb0027
    35 19D4 Light Chain CDR1 RASESVDTYGNNFIH
    36 19D4 Light Chain CDR2 LASNLES
    37 19D4 Light Chain CDR3 QQNNGDPWT
    38 19D4 Light Chain Variable Domain
    Figure imgb0028
    39 19D4 Heavy Chain CDR1 HPYMH
    40 19D4 Heavy Chain CDR2 RIDPANGNTKYDPKFQG
    41 19D4 Heavy Chain CDR3 EEVADYTMDY
    42 19D4 Heavy Chain Variable Domain
    Figure imgb0029
    43 24F12 Light Chain CDR1 SASQGINNFLN
    44 24F12 Light Chain CDR2 YTSSLHS
    45 24F12 Light Chain CDR3 QHFSKLPWT
    46 24F12 Light Chain Variable Domain
    Figure imgb0030
    47 24F12 Heavy Chain CDR1 SYAMS
    48 24F12 Heavy Chain CDR2 EIGSGGSYTYYPDTVTG
    49 24F12 Heavy Chain CDR3 ETTAGYFDY
    50 24F12 Heavy Chain
    Figure imgb0031
    Variable Domain WGQGTTLTVSS
    51 26B3 Light Chain CDR1 RTSENIFSYLA
    52 26B3 Light Chain CDR2 NAKTLAE
    53 26B3 Light Chain CDR3 QHHYAFPWT
    54 26B3 Light Chain Variable Domain
    Figure imgb0032
    55 26B3 Heavy Chain CDR1 GYFMN
    56 26B3 Heavy Chain CDR2 RIFPYNGDTFYNQKFKG
    57 26B3 Heavy Chain CDR3 GTHYFDY
    58 26B3 Heavy Chain Variable Domain
    Figure imgb0033
    59 9F3 Light Chain CDR1 AGGGCCAGCTCAACTGTAAGTTACAGTTACTTGCAC
    60 9F3 Light Chain CDR2 GGCACATCCAACTTGGCTTCT
    61 9F3 Light Chain CDR3 CAGCAGTACAGTGGTTACCCACTCACG
    62 9F3 Light Chain Variable Domain
    Figure imgb0034
    63 9F3 Heavy Chain CDR1 AGTGGTTATTACTGGAAC
    64 9F3 Heavy Chain CDR2 TACATAAAGTCCGACGGTAGCAATAATTACAACCCATCTCTCAAAAAT
    65 9F3 Heavy Chain CDR3 GAGTGGAAGGCTATGGACTAC
    66 9F3 Heavy Chain Variable Domain
    Figure imgb0035
    67 19D4 Light Chain CDR1 AGAGCCAGTGAAAGTGTTGATACTTATGGCAATAATTTTATACAC
    68 19D4 Light Chain CDR2 CTTGCATCCAACCTAGAATCT
    69 19D4 Light Chain CDR3 CAGCAAAATAATGGGGATCCGTGGACG
    70 19D4 Light CCAGCTTCTTTGGCTGTGTCTCTAGGGCAGAGGGCCACCATATCCTGCA
    Chain Variable Domain
    Figure imgb0036
    71 19D4 Heavy Chain CDR1 CACCCCTATATGCAC
    72 19D4 Heavy Chain CDR2
    Figure imgb0037
    73 19D4 Heavy Chain CDR3 GAGGAGGTGGCGGACTATACTATGGACTAC
    74 19D4 Heavy Chain Variable Domain
    Figure imgb0038
    75 24F12 Light Chain CDR1 AGTGCAAGTCAGGGCATTAACAATTTTTTAAAC
    76 24F12 Light Chain CDR2 TACACATCAAGTTTACACTCA
    77 24F12 Light Chain CDR3 CAGCACTTTAGTAAGCTTCCGTGGACG
    78 24F12 Light Chain Variable Domain
    Figure imgb0039
    79 24F12 Heavy Chain CDR1 AGCTATGCCATGTCT
    80 24F12 Heavy Chain CDR2
    Figure imgb0040
    81 24F12 Heavy Chain CDR3 GAAACTACGGCGGGCTACTTTGACTAC
    82 24F12 Heavy Chain Variable Domain
    Figure imgb0041
    83 26B3 Light Chain CDR1 CGAACAAGTGAGAATATTTTCAGTTATTTAGCA
    84 26B3 Light Chain CDR2 AATGCAAAAACCTTAGCAGAG
    85 26B3 Light Chain CDR3 CAACATCATTATGCTTTTCCGTGGACG
    86 26B3 Light Chain CCAGCCTCCCTATCTGCATCTGTGGGAGAAACTGTCACCATCACATGTC
    Variable Domain
    Figure imgb0042
    87 26B3 Heavy Chain CDR1 GGCTACTTTATGAAC
    88 26B3 Heavy Chain CDR2 CGTATTTTTCCTTACAATGGTGATACTTTCTACAACCAGAAGTTCAAGG GC
    89 26B3 Heavy Chain CDR3 GGGACTCATTACTTTGACTAC
    90 26B3 Heavy Chain Variable Domain
    Figure imgb0043

Claims (15)

  1. A method of assessing whether a subject is afflicted with an FRα-expressing cancer, the method comprising
    determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a serum or plasma sample derived from said subject; and
    comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein a difference between the level of FRα in the urine sample derived from said subject and the level of FRα in the control sample is an indication that the subject is afflicted with an FRα-expressing cancer;
    wherein the level of FRα which is not bound to a cell in the sample derived from said subject is assessed by contacting the sample with an antibody that binds FRα.
  2. The method of claim 1, wherein the FRα-expressing cancer is selected from the group consisting of lung cancer, mesothelioma, ovarian cancer, renal cancer, brain cancer, cervical cancer, nasopharyngeal cancer, squamous cell carcinoma of the head and neck, endometrial cancer, breast cancer, bladder cancer, pancreatic cancer, bone cancer, pituitary cancer, colorectal cancer and medullary thyroid cancer.
  3. The method of claim 1, wherein the level of FRα is determined by contacting the sample with an antibody selected from the group consisting of:
    (a) an antibody that binds the same epitope as the MORAb-003 antibody;
    (b) an antibody comprising SEQ ID NO:1 (GFTFSGYGLS) as CDRH1, SEQ ID NO:2 (MISSGGSYTYYADSVKG) as CDRH2, SEQ ID NO:3 (HGDDPAWFAY) as CDRH3, SEQ ID NO:4 (SVSSSISSNNLH) as CDRL1, SEQ ID NO:5 (GTSNLAS) as CDRL2 and SEQ ID NO:6 (QQWSSYPYMYT) as CDRL3;
    (c) the MOV18 antibody;
    (d) an antibody that binds the same epitope as the MOV18 antibody;
    (e) the 548908 antibody;
    (f) an antibody that binds the same epitope as the 548908 antibody;
    (g) the 6D398 antibody;
    (h) an antibody that binds the same epitope as the 6D398 antibody;
    (i) an antibody that binds the same epitope as the 26B3 antibody;
    (j) an antibody comprising SEQ ID NO:55 (GYFMN) as CDRH1, SEQ ID NO:56 (RIFPYNGDTFYNQKFKG) as CDRH2, SEQ ID NO:57 (GTHYFDY) as CDRH3, SEQ ID NO:51 (RTSENIFSYLA) as CDRL1 , SEQ ID NO:52 (NAKTLAE) as CDRL2 and SEQ ID NO:53 (QHHYAFPWT) as CDRL3;
    (k) the 26B3 antibody;
    (l) an antibody that binds the same epitope as the 19D4 antibody;
    (m) an antibody comprising SEQ ID NO:39 (HPYMH) as CDRH1, SEQ ID NO:40 (RIDPANGNTKYDPKFQG) as CDRH2, SEQ ID NO:41 (EEVADYTMDY) as CDRH3, SEQ ID NO:35 (RASESVDTYGNNFIH) as CDRL1, SEQ ID NO:36 (LASNLES) as CDRL2 and SEQ ID NO:37 (QQNNGDPWT) as CDRL3;
    (n) the 19D4 antibody;
    (o) an antibody that binds the same epitope as the 9F3 antibody;
    (p) an antibody comprising SEQ ID NO:31 (SGYYWN) as CDRH1, SEQ ID NO:32 (YIKSDGSNNYNPSLKN) as CDRH2, SEQ ID NO:33 (EWKAMDY) as CDRH3, SEQ ID NO:27 (RASSTVSYSYLH) as CDRL1, SEQ ID NO:28 (GTSNLAS) as CDRL2 and SEQ ID NO:29 (QQYSGYPLT) as CDRL3;
    (q) the 9F3 antibody;
    (r) an antibody that binds the same epitope as the 24F12 antibody;
    (s) an antibody comprising SEQ ID NO:47 (SYAMS) as CDRH1, SEQ ID NO:48 (EIGSGGSYTYYPDTVTG) as CDRH2, SEQ ID NO:49 (ETTAGYFDY) as CDRH3, SEQ ID NO:43 (SASQGINNFLN) as CDRL1, SEQ ID NO:44 (YTSSLHS) as CDRL2 and SEQ ID NO:45 (QHFSKLPWT) as CDRL3;
    (t) the 24F12 antibody;
    (u) an antibody that comprises a variable region light chain selected from the group consisting of LK26HuVK (SEQ ID NO: 13); LK26HuVKY (SEQ ID NO: 14); LK26HuVKPW (SEQ ID NO: 15); and LK26HuVKPW,Y (SEQ ID NO: 16);
    (v) an antibody that comprises a variable region heavy chain selected from the group consisting of LK26HuVH (SEQ ID NO: 17); LK26HuVH FAIS,N (SEQ ID NO: 18); LK26HuVH SLF (SEQ ID NO: 19); LK26HuVH I,I (SEQ ID NO: 20); and LK26KOLHuVH (SEQ ID NO: 21);
    (w) an antibody that comprises the heavy chain variable region LK26KOLHuVH (SEQ ID NO: 21) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16);
    (x) an antibody that comprises the heavy chain variable region LK26HuVH SLF (SEQ ID NO: 19) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16); and
    (y) an antibody that comprises the heavy chain variable region LK26HuVH FAIS,N (SEQ ID NO: 18) and the light chain variable region LK26HuVKPW,Y (SEQ ID NO: 16).
  4. The method of claim 1, wherein the antibody is selected from the group consisting of a murine antibody, a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, a Fab, Fab'2, ScFv, SMIP, affibody, avimer, versabody, nanobody, and a domain antibody, optionally, wherein the antibody is labeled with a label selected from the group consisting of a radio-label, a biotin-label, a chromophore-label, a fluorophore-label, an ECL label and an enzyme-label.
  5. The method of claim 1, wherein the level of FRα is determined by using a technique selected from the group consisting of western blot analysis, radioimmunoassay, immunofluorimetry, immunoprecipitation, equilibrium dialysis, immunodiffusion, solution phase assay, electrochemiluminescence immunoassay (ECLIA) and ELISA assay.
  6. The method of claim 1, wherein the control sample comprises a standardized control level of FRα in a healthy subject.
  7. The method of claim 1, wherein (i) the sample is treated with guanidine prior to determining the level of FRα in the sample, (ii) the sample is diluted prior to determining the level of FRα in the sample, and/or the sample is ; centrifuged, vortexed, or both, prior to determining the level of FRα in the sample.
  8. The method of claim 1, wherein the level of FRα in the sample derived from said subject is assessed by contacting the urine sample with a pair of antibodies selected from the group consisting of
    (a) MOV18 antibody immobilized to a solid support and labeled MORAB-003 antibody,
    (b) 9F3 antibody immobilized to a solid support and labeled 24F12 antibody,
    (c) 26B3 antibody immobilized to a solid support and labeled 19D4 antibody, and
    (d) 9F3 antibody immobilized to a solid support and labeled 26B3 antibody.
  9. A method of assessing the progression of an FRα-expressing cancer in a subject afflicted with the FRα- expressing cancer, the method comprising
    determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from said subject, wherein the sample is serum or plasma; and
    comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, thereby assessing the progression of the FRα-expressing cancer in said subject;
    wherein the level of FRα which is not bound to a cell in the sample derived from said subject is assessed by contacting the sample with an antibody that binds FRα.
  10. A method of stratifying a subject afflicted with an FRα-expressing cancer into one of at least four cancer therapy grounds comprising:
    determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from said subject, wherein the sample is serum or plasma; and
    stratifying the subject into one of at least four cancer therapy groups based on the level of folate receptor alpha (FRα) which is not bound to a cell;
    wherein the level of FRα which is not bound to a cell in the sample derived from said subject is assessed by contacting the sample with an antibody that binds FRα.
  11. The method of claim 10, wherein the FRα-expressing cancer is ovarian cancer and the subject is stratified in Stage I, Stage II, Stage III or Stage IV ovarian cancer.
  12. A method of monitoring the efficacy of MORAb-003 treatment of ovarian cancer or lung cancer in a subject suffering from ovarian cancer or lung cancer, the method comprising
    determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from said subject, wherein said subject has been previously administered MORAb-003 and wherein the sample is serum or plasma; and
    comparing the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from said subject with the level of FRα in a control sample, thereby monitoring the efficacy of MORAb-003 treatment.
  13. A method for predicting whether a subject suffering from ovarian cancer or lung cancer will respond to treatment with MORAb-003, the method comprising
    determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from said subject, wherein the sample is serum or plasma; and
    comparing the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from said subject with the level of FRα in a control sample,
    wherein a difference between the level of FRα in the sample derived from said subject and the level of FRα in the control sample is an indication as to whether the subject will respond to treatment with MORAb-003.
  14. A method of treating a subject having ovarian cancer or lung cancer, the method comprising
    determining the level of folate receptor alpha (FRα) which is not bound to a cell, in a sample derived from said subject, wherein the sample comprises serum or plasma; and
    comparing the level of folate receptor alpha (FRα) which is not bound to a cell with the level of FRα in a control sample, wherein a difference between the level of FRα in the sample derived from said subject and the level of FRα in the control sample is an indication that the subject is afflicted with ovarian cancer or lung cancer; and administering a therapeutically effective amount of MORAb-003 to said subject, thereby treating the subject having ovarian cancer or lung cancer.
  15. A kit for assessing whether a subject is afflicted with an FRα-expressing cancer or for assessing the progression of an FRα-expressing cancer in a subject, the kit comprising
    means for determining the level of folate receptor alpha (FRα) which is not bound to a cell in a sample derived from said subject; and
    instructions for use of the kit to assess whether the subject is afflicted with an FRα-expressing cancer or to assess the progression of an FRα-expressing cancer.
EP16186634.8A 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers Active EP3130605B1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US41049710P 2010-11-05 2010-11-05
US201161508444P 2011-07-15 2011-07-15
EP11785862.1A EP2635304B1 (en) 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers
PCT/US2011/059411 WO2012061759A2 (en) 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP11785862.1A Division-Into EP2635304B1 (en) 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers
EP11785862.1A Division EP2635304B1 (en) 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers

Publications (2)

Publication Number Publication Date
EP3130605A1 true EP3130605A1 (en) 2017-02-15
EP3130605B1 EP3130605B1 (en) 2020-01-08

Family

ID=45003076

Family Applications (2)

Application Number Title Priority Date Filing Date
EP11785862.1A Active EP2635304B1 (en) 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers
EP16186634.8A Active EP3130605B1 (en) 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP11785862.1A Active EP2635304B1 (en) 2010-11-05 2011-11-04 Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers

Country Status (14)

Country Link
US (1) US20120207771A1 (en)
EP (2) EP2635304B1 (en)
JP (2) JP6224456B2 (en)
KR (2) KR101931936B1 (en)
CN (2) CN103687618A (en)
AU (3) AU2011323124C1 (en)
BR (1) BR112013011072B1 (en)
CA (1) CA2816991C (en)
ES (2) ES2780192T3 (en)
IL (2) IL225871B (en)
MX (2) MX343607B (en)
RU (2) RU2630608C2 (en)
SG (3) SG10201509145XA (en)
WO (1) WO2012061759A2 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2172487A1 (en) * 2005-04-22 2010-04-07 Morphotek Inc. Antibodies with Immune Effector Activity that Internalize in Folate Receptor Alpha-Positive Cells
SG10201501342UA (en) 2010-02-24 2015-04-29 Immunogen Inc Folate receptor 1 antibodies and immunoconjugates and uses thereof
US20120189620A1 (en) * 2011-01-21 2012-07-26 Emory University Methods for treating non-functioning pituitary adenoma
EP2680839A1 (en) * 2011-03-02 2014-01-08 Morphotek, Inc. Co -administration of eribulin and farletuzumab for the treatment of breast cancer
EP3636279B1 (en) 2011-04-01 2023-09-13 ImmunoGen, Inc. Methods for increasing efficacy of folr1 cancer therapy
CN103998467B (en) * 2011-07-15 2016-12-14 卫材R&D管理有限公司 Anti-folacin receptor Alpha antibodies and application thereof
ES2766836T3 (en) 2012-05-15 2020-06-15 Eisai Inc Gastric Cancer Treatment Methods
CN104755498B (en) 2012-08-31 2019-06-18 伊缪诺金公司 For detecting the diagnostic assay and kit of folacin receptor 1
US20160311921A1 (en) * 2013-06-20 2016-10-27 Morphotek ,Inc. Methods for treatment of ovarian cancer
CN113150145A (en) 2013-08-30 2021-07-23 伊缪诺金公司 Antibodies and assays for detecting folate receptor 1
US20150132323A1 (en) * 2013-10-08 2015-05-14 Immunogen, Inc. Anti-FOLR1 Immunoconjugate Dosing Regimens
KR20240024318A (en) 2014-11-20 2024-02-23 에프. 호프만-라 로슈 아게 Combination therapy of t cell activating bispecific antigen binding molecules cd3 abd folate receptor 1 (folr1) and pd-1 axis binding antagonists
CA2983126A1 (en) 2015-04-17 2016-10-20 Morphotek, Inc. Methods for treating lung cancer
CA2994888A1 (en) 2015-09-17 2017-03-23 Immunogen, Inc. Therapeutic combinations comprising anti-folr1 immunoconjugates
JP7227123B2 (en) 2016-08-12 2023-02-21 エル.イー.エー.エフ. ホールディングス グループ エルエルシー α and γ-D polyglutamic acid antifolates and uses thereof
US10822410B2 (en) 2016-11-23 2020-11-03 Eisai R&D Management Co., Ltd. Anti-folate receptor alpha antibodies and uses thereof
CN107353325B (en) * 2017-07-26 2020-08-11 中国药科大学 Folate receptor alpha specific binding peptide 1 and application thereof
CN107446021B (en) * 2017-07-26 2020-08-11 中国药科大学 Folate receptor alpha specific binding peptide 5 and application thereof
CN107446020B (en) * 2017-07-28 2019-10-01 中国药科大学 Folacin receptor alpha specific peptide 2 and its application
AU2018328187A1 (en) * 2017-09-05 2020-03-26 Immunogen, Inc. Methods for detection of folate receptor 1 in a patient sample
JP2021512895A (en) 2018-02-07 2021-05-20 エル.イー.エー.エフ. ホールディングス グループ エルエルシーL.E.A.F. Holdings Group Llc Alpha polyglutamine oxidized pemetrexed and its use
EP3749316A4 (en) 2018-02-07 2021-10-27 L.E.A.F Holdings Group LLC Alpha polyglutamated pralatrexate and uses thereof
US11771700B2 (en) 2018-02-14 2023-10-03 L.E.A.F. Holdings Group Llc Gamma polyglutamated lometrexol and uses thereof
CN111954681B (en) 2018-03-13 2022-12-06 东莞凡恩世生物医药有限公司 Anti-folate receptor 1 antibodies and uses thereof
JP2022523564A (en) 2019-03-04 2022-04-25 アイオーカレンツ, インコーポレイテッド Data compression and communication using machine learning
US11396543B2 (en) 2019-04-29 2022-07-26 Immunogen, Inc. Biparatopic FR-α antibodies and immunoconjugates
KR20240017024A (en) * 2021-06-04 2024-02-06 이뮤노젠 아이엔씨 Treatment of cancer in patients with soluble FR-alpha
CN117700557A (en) * 2024-02-05 2024-03-15 卡秋(江苏)生物科技有限公司 Antibody or antigen binding fragment specifically binding to folate receptor alpha

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
WO1994004678A1 (en) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulins devoid of light chains
US5547835A (en) 1993-01-07 1996-08-20 Sequenom, Inc. DNA sequencing by mass spectrometry
US5605798A (en) 1993-01-07 1997-02-25 Sequenom, Inc. DNA diagnostic based on mass spectrometry
US5622824A (en) 1993-03-19 1997-04-22 Sequenom, Inc. DNA sequencing by mass spectrometry via exonuclease degradation
US5646253A (en) 1994-03-08 1997-07-08 Memorial Sloan-Kettering Cancer Center Recombinant human anti-LK26 antibodies
WO1998031700A1 (en) 1997-01-21 1998-07-23 The General Hospital Corporation Selection of proteins using rna-protein fusions
US6261804B1 (en) 1997-01-21 2001-07-17 The General Hospital Corporation Selection of proteins using RNA-protein fusions
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2004113388A2 (en) 2003-05-23 2004-12-29 Morphotek, Inc. Anti alpha - folate - receptor - tetramer antibodies
US20050232919A1 (en) 2004-02-12 2005-10-20 Morphotek, Inc. Monoclonal antibodies that specifically block biological activity of a tumor antigen
US7115396B2 (en) 1998-12-10 2006-10-03 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2009132081A2 (en) 2008-04-24 2009-10-29 The Research Foundation Of State University Of New York Monoclonal antibody-based targeting of folate receptors
US7754698B2 (en) 2007-01-09 2010-07-13 Isis Pharmaceuticals, Inc. Modulation of FR-alpha expression

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6933108B2 (en) * 1999-12-09 2005-08-23 The Regents Of The University Of California Methods and compositions for use in the treatment of filovirus mediated disease conditions
JP2004512540A (en) * 2000-11-01 2004-04-22 マウント・サイナイ・ホスピタル Ovarian cancer detection
US20100120078A1 (en) * 2001-08-16 2010-05-13 Tony Baker Urine Stabilization System
CA2784450C (en) * 2001-11-13 2016-09-13 Id Biomedical Corporation Polypeptides of pseudomonas aeruginosa
AU2003253589A1 (en) * 2002-03-26 2003-11-10 Centocor, Inc. Epitope mapping using nuclear magnetic resonance
US20080131366A1 (en) * 2003-03-17 2008-06-05 Manohar Ratnam Folate Receptor Gene Modulation For Cancer Diagnosis And Therapy
US20080113344A1 (en) * 2003-10-28 2008-05-15 Ralph Wirtz Methods and Compositions for the Response Prediction of Malignant Neoplasia to Treatment
AU2006230219A1 (en) * 2005-03-30 2006-10-05 Endocyte, Inc. Method for cancer prognosis using cellular folate vitamin receptor quantification
EP2172487A1 (en) * 2005-04-22 2010-04-07 Morphotek Inc. Antibodies with Immune Effector Activity that Internalize in Folate Receptor Alpha-Positive Cells
CN101384903A (en) * 2006-02-09 2009-03-11 南佛罗里达大学 Detection of cancer by elevated levels of bcl-2
US8486412B2 (en) * 2006-06-01 2013-07-16 Mayo Foundation For Medical Education And Research Immunity to folate receptors
EP1900752A1 (en) * 2006-09-15 2008-03-19 DOMPE' pha.r.ma s.p.a. Human anti-folate receptor alpha antibodies and antibody fragments for the radioimmunotherapy of ovarian carcinoma
AU2008317404B2 (en) * 2007-10-19 2014-12-18 Cell Signaling Technology, Inc. Cancer classification and methods of use
US20110177525A1 (en) * 2010-01-19 2011-07-21 Predictive Biosciences, Inc. Antibodies and methods of diagnosing diseases
JP2013540995A (en) * 2010-08-18 2013-11-07 カリス ライフ サイエンシズ ルクセンブルク ホールディングス エス.アー.エール.エル. Circulating biomarkers for disease

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5759808A (en) 1992-08-21 1998-06-02 Vrije Universiteit Brussel Immunoglobulins devoid of light chains
WO1994004678A1 (en) 1992-08-21 1994-03-03 Casterman Cecile Immunoglobulins devoid of light chains
US5547835A (en) 1993-01-07 1996-08-20 Sequenom, Inc. DNA sequencing by mass spectrometry
US5605798A (en) 1993-01-07 1997-02-25 Sequenom, Inc. DNA diagnostic based on mass spectrometry
US5622824A (en) 1993-03-19 1997-04-22 Sequenom, Inc. DNA sequencing by mass spectrometry via exonuclease degradation
US6124106A (en) 1994-03-08 2000-09-26 Ludwig Institute For Cancer Research Method for detecting cancers
US5646253A (en) 1994-03-08 1997-07-08 Memorial Sloan-Kettering Cancer Center Recombinant human anti-LK26 antibodies
WO1998031700A1 (en) 1997-01-21 1998-07-23 The General Hospital Corporation Selection of proteins using rna-protein fusions
US6258558B1 (en) 1997-01-21 2001-07-10 The General Hospital Corporation Method for selection of proteins using RNA-protein fusions
US6261804B1 (en) 1997-01-21 2001-07-17 The General Hospital Corporation Selection of proteins using RNA-protein fusions
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US7115396B2 (en) 1998-12-10 2006-10-03 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2004113388A2 (en) 2003-05-23 2004-12-29 Morphotek, Inc. Anti alpha - folate - receptor - tetramer antibodies
US20050232919A1 (en) 2004-02-12 2005-10-20 Morphotek, Inc. Monoclonal antibodies that specifically block biological activity of a tumor antigen
US7754698B2 (en) 2007-01-09 2010-07-13 Isis Pharmaceuticals, Inc. Modulation of FR-alpha expression
WO2009132081A2 (en) 2008-04-24 2009-10-29 The Research Foundation Of State University Of New York Monoclonal antibody-based targeting of folate receptors

Non-Patent Citations (83)

* Cited by examiner, † Cited by third party
Title
"Guide to Protein Purification", vol. 182, 1990, ACADEMIC PRESS, INC., article "Methods in Enzymology"
ADAM, B.L. ET AL., CANCER RES, vol. 62, 2002, pages 3609
BIBBO ET AL., ACTA. CYTOL, vol. 46, 2002, pages 25 - 29
BIBBO ET AL., ANAL. QUANT. CYTOL. HISTOL., vol. 25, 2003, pages 8 - 11
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BUENO R ET AL., J OF THORACIC AND CARDIOVASCULAR SURGERY, vol. 121, no. 2, 2001, pages 225 - 233
CARELL ET AL., ANGEW. CHEM. INT. ED. ENGL, vol. 33, 1994, pages 2061
CARRELL ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 33, 1994, pages 2059
CHO ET AL., SCIENCE, vol. 261, 1993, pages 1303
CONEY ET AL., CANCER RES, vol. 51, 1991, pages 6125 - 6132
CORTEZ-RETAMOZO ET AL., INT. J. CANCER, vol. 89, 2002, pages 456 - 62
CULL ET AL., PROC NATL ACAD SCI USA, vol. 89, 1992, pages 1865 - 1869
CWIRLA ET AL., PROC. NATL. ACAD. SCI., vol. 87, 1990, pages 6378 - 6382
D K ARMSTRONG ET AL: "Exploratory phase II efficacy study of MORAb-003, a monoclonal antibody against folate receptor alpha, in platinum-sensitive ovarian cancer in first relapse.", J CLIN ONCOL MAY 20 SUPPL; ABSTR 5500, 1 January 2008 (2008-01-01), XP055022660, Retrieved from the Internet <URL:http://hwmaint.meeting.ascopubs.org/cgi/content/abstract/26/15_suppl/5500> [retrieved on 20120322] *
DEVLIN, SCIENCE, vol. 249, 1990, pages 404 - 406
DEWITT ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 90, 1993, pages 6909
DOLO V ET AL: "Matrix-degrading proteinases are shed in membrane vesicles by ovarian cancer cells in vivo and in vitro", CLINICAL AND EXPERIMENTAL METASTASIS, SPRINGER NETHERLANDS, NL, vol. 17, no. 2, 1 March 1999 (1999-03-01), pages 131 - 140, XP002461206, ISSN: 0262-0898, DOI: 10.1023/A:1006500406240 *
DUMOULIN ET AL., NATURE, vol. 424, 2003, pages 783 - 788
EATI BASAL ET AL: "Functional Folate Receptor Alpha Is Elevated in the Blood of Ovarian Cancer Patients", PLOS ONE, vol. 4, no. 7, 20 July 2009 (2009-07-20), pages e6292, XP055022613, DOI: 10.1371/journal.pone.0006292 *
ELKANAT H; RATNAM M., FRONTIERS IN BIOSCIENCE, vol. 11, 2006, pages 506 - 519
ERB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 11422
FELICI, J. MOL. BIOL., vol. 222, 1991, pages 301 - 310
FISHER R.E., J NUCL MED, vol. 49, 2008, pages 899 - 906
FODOR, NATURE, vol. 364, 1993, pages 555 - 556
FRANKLIN, WA ET AL., INT J CANCER, vol. 8, 1994, pages 89 - 95
FRANKLIN, WA ET AL., INT J CANCER,, vol. 8, 1994, pages 89 - 95
GALLOP ET AL., J. MED. CHEM., vol. 37, 1994, pages 1233
GREENE FL, PAGE DL, FLEMING ID, FRITZ AG, BALCH CM, HALLER DG, ET AL.,: "AJCC Cancer Staging Manual. 6th edition", 2002, SPRINGER-VERLAG, pages: 167 - 77
HARLOW AND LANE,: "Antibodies, A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
HARLOW; LANE: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
HARTMAN L.C. ET AL., INT J CANCER, vol. 121, 2007, pages 938 - 942
HILL, T; LEWICKI, P.: "STATISTICS Methods and Applications", STATSOFT, 2006
HOLLIGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOLLIGER; WINTER, CANCER IMMUNOL. IMMUNOTHER., vol. 45, no. 3-4, 1997, pages 128 - 30
HOLLIGER; WINTER, CANCER IMMUNOL. IMMUNOTHER., vol. 45, no. 34, 1997, pages 128 - 30
HOUGHTEN, BIOTECHNIQUES, vol. 13, 1992, pages 412 - 421
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
IWAKIRI S ET AL., ANNALS OF SURGICAL ONCOLOGY,, vol. 15, no. 3, pages 889 - 899
KOSTELNY ET AL., J. IMMUNOL., vol. 148, 1992, pages 1547 - 1553
LAM, ANTICANCER DRUG DES., vol. 12, 1997, pages 145
LAM, NATURE, vol. 354, 1991, pages 82 - 84
LARONGA, C. ET AL., DIS MARKERS, vol. 19, 2003, pages 229
LAUWEREYS ET AL., EMBO J., vol. 17, 1998, pages 3512 - 3520
LI, J ET AL., CLIN CHEM, vol. 48, 2002, pages 1296
LI, J. ET AL., CLIN CHEM, vol. 48, 2002, pages 1296
LINDA E. KELEMEN: "The role of folate receptor [alpha] in cancer development, progression and treatment: Cause, consequence or innocent bystander?", INTERNATIONAL JOURNAL OF CANCER, vol. 119, no. 2, 1 January 2006 (2006-01-01), pages 243 - 250, XP055027169, ISSN: 0020-7136, DOI: 10.1002/ijc.21712 *
LU ET AL., J. BIOL. CHEM., vol. 279, no. 4, 2004, pages 2856 - 65
MANTOVANI L T ET AL: "Folate binding protein distribution in normal tissues and biological fluids from ovarian carcinoma patients as detected by the monoclonal antibodies MOv18 and MOv19", EUROPEAN JOURNAL OF CANCER, ELSEVIER, AMSTERDAM, NL, vol. 30, no. 3, 1 January 1994 (1994-01-01), pages 363 - 369, XP022643304, ISSN: 0959-8049, [retrieved on 19940101], DOI: 10.1016/0959-8049(94)90257-7 *
MIOTTI, S. ET AL., INT J CANCER, vol. 38, 1987, pages 297 - 303
NAMBA ET AL., ANALYTICAL SCIENCE, vol. 15, 1999, pages 1087 - 1093
PARKER N. ET AL., ANALYTICAL BIOCHEMISTRY, vol. 338, 2005, pages 284 - 293
PETRICOIN, E.F ET AL., DIS MARKERS, vol. 359, 2002, pages 572
PETRICOIN, E.F. ET AL., DIS MARKERS, vol. 359, 2002, pages 572
PHILIP S LOW ET AL: "Discovery and Development of Folic-Acid-Based Receptor Targeting for Imaging and Therapy of Cancer and Inflammatory Diseases", ACCOUNTS OF CHEMICAL RESEARCH, ACS, WASHINGTON, DC, US, vol. 41, no. 1, 1 January 2008 (2008-01-01), pages 120 - 129, XP007915422, ISSN: 0001-4842, [retrieved on 20070727], DOI: 10.1021/AR7000815 *
PLESCHBERGER ET AL., BIOCONJUGATE CHEM, vol. 14, 2003, pages 440 - 448
PLUCKTHUN; PACK, IMMUNOTECHNOLOGY, vol. 3, no. 2, 1997, pages 83 - 105
POLJAK ET AL., STRUCTURE, vol. 2, 1994, pages 1121 - 1123
PRICE AND NEWMAN,: "Principles And Practice Of Immunoassay, 2nd Edition,", 1997, MACMILLAN
R. SCOPES: "Protein Purification", 1982, SPRINGER-VERLAG
RICHART ET AL., AM. J. OBSTET. GYNECOL., vol. 105, 1969, pages 386
RIDGWAY ET AL., PROTEIN ENG., vol. 9, no. 7, 1996, pages 617 - 21
ROBERTS; SZOSTAK, PROC. NATL. ACAD. SCI USA, vol. 94, 1997, pages 12297
SABA N.F. ET AL., HEAD NECK, vol. 31, no. 4, 2009, pages 475 - 481
SAQI ET AL., DIAGN. CYTOPATHOL, vol. 27, 2003, pages 365 - 370
SCOTT; SMITH, SCIENCE, vol. 249, 1990, pages 386 - 390
SCOUTEN, W. H., METHODS IN ENZYMOLOGY, vol. 135, 1987, pages 30 - 65
SOBIN LH, WITTEKIND CH: "TNM classification of malignant tumours. 6th edition.", 2002, WILEY-LISS, article "International Union Against Cancer"
SONGSIVILAI; LACHMANN, CLIN. EXP. IMMUNOL., vol. 79, 1990, pages 315 - 321
SPIRA J; ETTINGER, D.S.: "Multidisciplinary management of lung cancer", N ENGL J MED, vol. 350, 2004, pages 382
STIJLEMANS ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 1256 - 1261
THUYET TRAN ET AL: "Enhancement of Folate Receptor A Expression in Tumor Cells Through the Glucocorticoid Receptor: A Promising Means to Improved Tumor Detection and Targeting", CANCER RES, vol. 65, no. 10, 15 May 2005 (2005-05-15), pages 4431 - 4441, XP055027294 *
TOLSON, J. ET AL., LAB INVEST, vol. 84, 2004, pages 845
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
WEIR, D. M.: "Handbook of Experimental Immunology", 1986, BLACKWELL SCIENTIFIC
WEITMAN SD ET AL., CANCER RES, vol. 52, pages 6708 - 6711
WEITMAN, SD ET AL., CANCER RES, vol. 52, 1992, pages 3396 - 3401
WRIGHT, G.L., JR. ET AL., EXPERT REV MOL DIAGN, vol. 2, 2002, pages 549
WU ET AL., IMMUNOTECHNOLOGY, vol. 2, no. 1, 1996, pages 21 - 36
WU ET AL., IMMUNOTECHNOLOGY, vol. 2, no. L, 1996, pages 21 - 36
XIAO, Z. ET AL., CANCER RES, vol. 61, 2001, pages 6029
YANG R ET AL., CLIN CANCER RES, vol. 13, 2007, pages 2557 - 2567
ZUCKERMANN ET AL., J. MED. CHEM., vol. 37, 1994, pages 2678
ZUCKERMANN ET AL., J. MED. CHEM., vol. 37, 1994, pages 2678 - 85

Also Published As

Publication number Publication date
JP2018036270A (en) 2018-03-08
RU2013125772A (en) 2014-12-10
AU2011323124C1 (en) 2016-09-22
SG189957A1 (en) 2013-06-28
MX343607B (en) 2016-11-11
KR102057438B1 (en) 2019-12-20
ES2627061T3 (en) 2017-07-26
KR20180137587A (en) 2018-12-27
SG10201701933XA (en) 2017-05-30
WO2012061759A3 (en) 2012-07-26
JP2013544354A (en) 2013-12-12
EP2635304A2 (en) 2013-09-11
JP6224456B2 (en) 2017-11-01
AU2018201760A1 (en) 2018-04-05
AU2011323124B2 (en) 2016-02-25
JP6554152B2 (en) 2019-07-31
KR101931936B1 (en) 2018-12-26
RU2764592C2 (en) 2022-01-18
BR112013011072B1 (en) 2021-02-02
AU2018201760B2 (en) 2019-07-25
RU2017129266A (en) 2019-02-04
EP2635304B1 (en) 2017-03-08
CA2816991C (en) 2020-06-09
IL225871A0 (en) 2013-06-27
SG10201509145XA (en) 2015-12-30
MX368394B (en) 2019-10-01
AU2016203408A1 (en) 2016-06-16
RU2630608C2 (en) 2017-09-11
EP3130605B1 (en) 2020-01-08
CA2816991A1 (en) 2012-05-10
ES2780192T3 (en) 2020-08-24
KR20140024838A (en) 2014-03-03
IL225871B (en) 2018-08-30
CN108562744A (en) 2018-09-21
US20120207771A1 (en) 2012-08-16
MX2013005067A (en) 2013-07-02
BR112013011072A2 (en) 2017-06-20
IL260590B (en) 2020-01-30
CN103687618A (en) 2014-03-26
AU2011323124A1 (en) 2013-05-09
RU2017129266A3 (en) 2021-01-25
WO2012061759A2 (en) 2012-05-10

Similar Documents

Publication Publication Date Title
EP3130605B1 (en) Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha-expressing cancers
JP5711196B2 (en) Use of HE4 and other biochemical markers to determine ovarian cancer
JP7174385B2 (en) Use of Laminin-2 to Diagnose Pancreatic Cancer
US8252904B2 (en) Glycodelin monoclonal antibodies and methods for their use in the detection of ovarian cancer
KR20090007454A (en) Use of protein s100a12 as a marker for colorectal cancer
EP3102600B1 (en) Composition and method for detecting malignant neoplastic disease
KR20110091423A (en) Composition for diagnosing susceptibility to cancer comprising anti-tmap/ckap2 antibody

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 2635304

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20170811

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20180126

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: EISAI INC.

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20190617

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AC Divisional application: reference to earlier application

Ref document number: 2635304

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011064586

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1222607

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200215

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: HEPP WENGER RYFFEL AG, CH

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200531

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200408

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2780192

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20200824

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200408

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200508

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200409

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602011064586

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1222607

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200108

26N No opposition filed

Effective date: 20201009

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201104

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20201130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201104

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200108

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201130

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230520

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20231126

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231127

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20231201

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20231127

Year of fee payment: 13

Ref country code: IT

Payment date: 20231122

Year of fee payment: 13

Ref country code: FR

Payment date: 20231127

Year of fee payment: 13

Ref country code: DE

Payment date: 20231129

Year of fee payment: 13

Ref country code: CH

Payment date: 20231201

Year of fee payment: 13